Seven sirtuins for seven deadly diseases ofaging

Seven sirtuins for seven deadly diseases ofaging

Free Radical Biology and Medicine 56 (2013) 133–171 Contents lists available at SciVerse ScienceDirect Free Radical Biology and Medicine journal hom...

2MB Sizes 3 Downloads 122 Views

Free Radical Biology and Medicine 56 (2013) 133–171

Contents lists available at SciVerse ScienceDirect

Free Radical Biology and Medicine journal homepage: www.elsevier.com/locate/freeradbiomed

Review Article

Seven sirtuins for seven deadly diseases of aging Brian J. Morris n Basic & Clinical Genomics Laboratory, School of Medical Sciences and Bosch Institute, Building F13, University of Sydney, NSW 2006, Australia

a r t i c l e i n f o

abstract

Article history: Received 25 June 2012 Received in revised form 31 August 2012 Accepted 5 October 2012 Available online 24 October 2012

Sirtuins are a class of NAD þ -dependent deacetylases having beneficial health effects. This extensive review describes the numerous intracellular actions of the seven mammalian sirtuins, their protein targets, intracellular localization, the pathways they modulate, and their role in common diseases of aging. Selective pharmacological targeting of sirtuins is of current interest in helping to alleviate global disease burden. Since all sirtuins are activated by NAD þ , strategies that boost NAD þ in cells are of interest. While most is known about SIRT1, the functions of the six other sirtuins are now emerging. Best known is the involvement of sirtuins in helping cells adapt energy output to match energy requirements. SIRT1 and some of the other sirtuins enhance fat metabolism and modulate mitochondrial respiration to optimize energy harvesting. The AMP kinase/SIRT1–PGC-1a–PPAR axis and mitochondrial sirtuins appear pivotal to maintaining mitochondrial function. Downregulation with aging explains much of the pathophysiology that accumulates with aging. Posttranslational modifications of sirtuins and their substrates affect specificity. Although SIRT1 activation seems not to affect life span, activation of some of the other sirtuins might. Since sirtuins are crucial to pathways that counter the decline in health that accompanies aging, pharmacological agents that boost sirtuin activity have clinical potential in treatment of diabetes, cardiovascular disease, dementia, osteoporosis, arthritis, and other conditions. In cancer, however, SIRT1 inhibitors could have therapeutic value. Nutraceuticals such as resveratrol have a multiplicity of actions besides sirtuin activation. Their net health benefit and

Keywords: Life span AMP kinase PGC-1a Mitochondria Oxidative phosphorylation Resveratrol Reactive oxygen species Intracellular mechanisms Pharmacotherapy Molecular genetics Aging Cardiovascular disease

Abbreviations: ABCA1, ATP-binding cassette transporter A1; ACADL, acyl-CoA dehydrogenase, long chain; ACSS2, acyl-CoA synthase short-chain family member 2; AgRP, agouti-related peptide; AKT, also known as protein kinase B; ALDH2, aldehyde dehydrogenase 2 family, mitochondrial; AMPK, AMP kinase; APC, anaphase-promoting complex; AROS, activator of SIRT1; ATG, autophagy-related protein; ATF4, activating transcription factor 4; ATM, ataxia telangiectasia mutated; ATP5A1, F1F0-ATP synthase, H þ transporting, mitochondrial F1 complex, a subunit 1; BATF, basic leucine zipper transcription factor; BCL6, B cell lymphoma 6 protein; BCR, breakpoint cluster region gene; BMAL1, brain and muscle ARNT-like 1; CBP, CREB-binding protein; CCAR, cell cycle and apoptosis regulator; CDK, cyclin-dependent kinase; CHK, cellcycle checkpoint kinase; ChREBP, carbohydrate responsive element-binding protein; CIITA, class II transactivator; CNS, central nervous system; CoA, coenzyme A; COX, cyclooxygenase; CPS1, carbamoyl phosphate synthetase 1; CPT1, carnitine palmitoyl transferase-1; CRTC, CREB-regulated transcription coactivator; CREB, cyclic-AMPresponsive-element-binding protein; CreP, constitutive repressor of eIF2a phosphorylation; CRP, C-reactive protein; DBC1, deleted in breast cancer 1; DNMT1, DNA (cytosine-5)-methyltransferase 1; DYRK, dual specificity tyrosine-phosphorylated and regulated kinase; E2F1, E2F transcription factor 1; eIF2a, eukaryotic initiation factor 2a; EPC, endothelial progenitor cell; eNOS, endothelial nitric oxide synthase; ER, estrogen receptor; ERK1/2, extracellular signal-regulated kinase 1/2; EVI1, ectopic viral integration site I; FMRP, fragile X mental retardation protein; FOXO, forkhead box; class O, transcription factors; FXR, farnesoid X receptor; G6P, glucose 6-phosphatase; GADD34, growth arrest and DNA damage-inducible protein 34; GDH, glutamate dehydrogenase; HBX, hepatitis B virus X protein; HIF, hypoxia-inducible factor; HISTH3, histone cluster 3, H3; HMGCS2, 3-hydroxy-3-methylglutaryl CoA synthase 2; HNF, hepatocyte nuclear factor 1; HOXA, homeobox A; HSF, heat shock factor; IDH2, isocitrate dehydrogenase 2; IGF-1, insulin-like growth factor-1; IKZF, ikaros family zinc finger protein; JNK, JUN N-terminal kinase; KU70, DNA repair factor KU70; LC3-II, microtubule-associated protein 1 light chain 3-II; LCAD, long-chain acyl CoA dehydrogenase; LKB1, liver kinase B1; LOX-1, lectin-like oxidized LDL receptor-1; LSD, methyltransferase lysine-specific demethylase; LXR, liver X receptor; MAP, mitogen-activated protein; MEF2, myocyte enhancing factor 2; MHC, major histocompatibility complex; MKP3, mitogen activated protein kinase phosphatase 3; MMP, matrix metalloproteinase; MRPL10, mitochondrial ribosomal protein L10; MYOD, myoblast determination protein; NAMPT, nicotinamide phosphororibosyltransferase; NBS1, Nijmegen Breakage Syndrome 1; NCoR, nuclear receptor corepressor; NDUFA9, NADH dehydrogenase (ubiquinone) 1a subcomplex 9; NDUFS8, NADH:ubiquinone oxidoreductase 8; NF-kB, nuclear factor-kB; NA, nicotinic acid; NMN, nicotinamide mononucleotide; NR, nicotinamide riboside; OTC, ornithine transcarbamoyltransferase; OXPHOS, oxidative phosphorylation; P450SCC, P450 side chain cleavage enzyme; p53, tumor suppressor p53; PAR3, partitioning defective 3 homologue; PARP, poly(ADP-ribose) polymerase; PEPCK, phosphoenolpyruvate carboxykinase; PER2, period circadian protein homolog 2; PGC1a, peroxisome proliferator-activated receptor-g coactivator 1a; Pi, inorganic phosphate; PID, peptidylpropyl isomerase D (cyclophilin D); PKA, cAMP-dependent protein kinase A; PKB, protein kinase B; POMC, proopiomelanocortin; PPAR, peroxisome proliferator-activated receptor; PRC, polycomb repressive group complex; PTEN, phosphatase and tensin homolog; RARb, retinoic acid receptor-b; Rb, retinoblastoma protein; RIP, receptor-interacting protein; ROS, reactive oxygen species; RUNX2, runt-related transcription factor 2; SDH, succinate dehydrogenase; SDHA, SDH subunit A flavoprotein; SIRT, sirtuin; SMRT, silencing mediator of retinoid and thyroid hormone receptor; SNP, single nucleotide polymorphism; SOD, superoxide dismutase; SOST, sclerostin; SREBP, sterol regulatory elementbinding protein; STAC, sirtuin activating compound; STAT3, signal transducer and activator of transcription 3; STAU, staufen; STK11, serine/threonine kinase 11 (LBK1); SUMO, small ubiquitin-like modifier; SUV39H1, a methyltransferase; TCA, tricarboxylic acid cycle; TDP-43, 43-kDa transactive response DNA-binding protein; TORC, CREBregulated transcription coactivator; UCP2, uncoupling protein 2; USP22, ubiquitin specific peptidase 22; WRN, Werner syndrome ATP-dependent helicase; XRCC6, X-ray repair complementing defective repair in Chinsese hamster cells 6 (Ku70) n Fax: þ61 2 9351 2227. E-mail address: [email protected] 0891-5849/$ - see front matter & 2012 Elsevier Inc. All rights reserved. http://dx.doi.org/10.1016/j.freeradbiomed.2012.10.525

134 Type 2 diabetes Obesity Dementia Cancer

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

relative safety may have originated from the ability of animals to survive environmental changes by utilizing these stress resistance chemicals in the diet during evolution. Each sirtuin forms a key hub to the intracellular pathways affected. & 2012 Elsevier Inc. All rights reserved.

Contents Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 134 Intracellular localization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 135 Enzymatic activity of each sirtuin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136 Actions of nuclear sirtuins (SIRT1 and SIRT6) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136 SIRT1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136 SIRT6 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 137 Actions of mitochondrial sirtuins (SIRT3, SIRT4, and SIRT5) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 138 SIRT3 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 138 SIRT4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 138 SIRT5 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 138 Actions of the cytosolic sirtuin (SIRT2) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 138 Actions of the nucleolar sirtuin (SIRT7) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 139 Transcriptional regulation of sirtuin genes. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 139 Posttranscriptional regulation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 139 Posttranslational regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 139 Formation of complexes with other proteins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 140 Roles for sirtuins in tissue functions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 140 Liver . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 140 Pancreas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 142 Muscle . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 142 Adipose tissue . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 142 Brain and nerves . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 142 Sirtuins in metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 144 Chemical compounds that activate sirtuins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 145 Obesity. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 148 Type 2 diabetes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 149 Cardiovascular disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 149 Neurodegenerative diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 151 Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 152 Does SIRT1 promote cancer?. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 152 An anticancer role for SIRT1? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 154 Anticancer drugs and SIRT1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 154 SIRT2 in cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 154 SIRT3 in cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 155 SIRT4 in cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 155 SIRT6 in cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 155 SIRT7 in cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 155 How might the dilemma be resolved? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 155 Inflammatory arthropathies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 155 Osteoporosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 156 Reproductive function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 156 Other medical conditions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 156 Aging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 156 Clinical molecular genetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 158 Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 158 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 158

Introduction The ‘‘Seven Deadly Sins’’ of old (sloth, gluttony, wrath, greed, pride, lust, and envy) have overlapping counterparts in modern society (sloth, gluttony, junk food, smoking, alcoholism, drug abuse, and psychosocial stress). These ‘‘sins’’ contribute to the seven deadly conditions that increase in prevalence with aging (obesity, type 2 diabetes, cardiovascular disease, cancer, dementia, arthritis, and osteoporosis). The sirtuins are a class of NAD þ -dependent deacetylases comprising seven members in humans and other mammals [1].

These enzymes have attracted major interest because of their apparent roles as protectors, and controversially, as contributors to all or some of the life-threatening conditions of aging. More specifically, the sirtuins are important in the transduction pathways emanating from energy sensing. Their ability to regulate systems that control the redox environment has the potential to help counteract oxidative damage that is associated with common diseases of aging and that contributes to aging itself [2]. Because sirtuin malfunction likely has pathophysiological consequences in common clinical conditions of aging pharmaceutical agents targeting sirtuins have been developed [3].

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

The history leading up to the explosion in research on sirtuins stems from the finding that calorie restriction can extend rat life span [4,5] and later evidence that the natural flavanoid resveratrol might mimic this effect in lower organisms [6–8]. The gain in life expectancy achieved by calorie restriction was suggested to exceed that achieved by curing cardiovascular disease, cancer, and type 2 diabetes combined [9]. Since population level calorie restriction is unrealistic, many therefore asked whether sirtuin activation might mimic calorie restriction [10] or at least delay onset of age-related diseases by ‘‘compression of morbidity’’ [11]. Although the first sirtuin was discovered in 1984 in yeast [12], interest did not really take off until an effect on life span was noted, first in yeast in 1997 [13], then in higher eukaryotes such as the nematode worm Caenorhabditis elegans in 2001 [14] and the fruit fly Drosophila melanogaster in 2004 [15]. The nature of sirtuins as NAD þ -dependent deacetylases was recognized in 2000 [16] and immediately implicated them in the metabolic state of the cell [17]. Once their enzymatic actions in the cell started to be elucidated it was soon realized that their apparent ability to extend life span involved similar pathways as utilized by calorie restriction. A role for the most-studied sirtuin, SIRT1, in life span has now been refuted, however. Manipulation of SIRT1 expression or activity has little or no effect on mammalian life span [18,19]. And subsequent studies in model organisms using better controls showed that SIRT1 activation or deletion had little [20,21] or no [22] effect on their life span. So too calorie restriction it seems, at least in primates, with the 25-year study of rhesus monkeys having found no effect on life span [23], in contrast to earlier findings by others that did [24]. The earlier study involved ad libitum feeding of controls with a different diet that notably included 28% (as opposed to 4%) sucrose, leading to overweight and diabetes. So earlier death of controls may partly explain the discrepancy. Disease onset was later and health markers were nevertheless more favorable in the calorie-restricted group of each cohort of monkeys. Overexpression of SIRT6, however, extended the life span of male mice by 15%, in effect extinguishing the sex difference in mouse longevity [25]. This effect was seen in mice with different genetic backgrounds. While SIRT1 deacetylates numerous targets, the best-known substrates for SIRT6 are two histones (H3K9 and HK56); the targeting of these explains SIRT6’s role in DNA damage response, especially during oxidative stress, and in telomere maintenance [26–28]. While SIRT1 is more similar structurally to the single sirtuin present in yeast, SIRT6 is more similar functionally to yeast sirtuin [29]. Male SIRT6 transgenic mice exhibit a female metabolic profile, most notably in fat tissue, in which

changes seem linked to life span [29]. The liver of male mice showed the biggest changes in gene expression [25], which overlapped those seen in calorie-restricted mice [30,31]. Serum IGF-1 level was reduced modestly in male mice, as was IGF-1 signaling in adipose tissue [25]. Thus SIRT6 has partial feminizing effects in male mice. Loss of SIRT6 causes severe metabolic defects, rapid aging, and death at 4 weeks of age [32]. The seven mammalian sirtuins all appear to be important in suppression of such common diseases of aging as cardiovascular disease, type 2 diabetes and dementia [33–35]. Effects involving mitochondrial function are crucial to this. Here I provide the most extensive review of sirtuins to date. This describes their function and roles in disease processes. Because there are over 2500 publications on sirtuins, only a selective overview is provided. Moreover, for simplicity, this review will use the human nomenclature ‘‘SIRT’’ rather than ‘‘Sirt’’ or ‘‘SirT’’ to refer to both the human and rodent sirtuin proteins and genes. All abbreviations are shown in the footnote on the previous page.

Intracellular localization SIRT1 is localized in the nucleus [36] (Table 1), but it shuttles to the cytoplasm when required to act on cytoplasmic targets, such as during inhibition of insulin signaling [37]. During prometaphase, levels increase and SIRT1 associates with mitotic chromatin until telophase [38]. It mediates loading of histone H1 and the condensing I complex to chromatin, thus contributing to chromosomal condensation [38]. In contrast, SIRT2 is cytoplasmic (Table 1). It deacetylates tubulin microtubules [39] and transcription factors that shuttle from the cytoplasm to the nucleus [40]. During mitosis SIRT2 is required for exit of cells from the mitotic phase [41]. It localizes to chromatin and, in part by a dynamic interplay with the human ortholog of MOF [42], deacetylates acetylated histone H4K16, and thus decreases the latter during G2/M transition [43]. SIRT2 is thereby pivotal to formation of condensed chromatin when the latter must be generated anew [43]. Moreover, SIRT2, by deacetylating BubR1 kinase, may help ensure faithful chromosome separation during mitosis [44]. SIRT3, SIRT4, and SIRT5 are located in mitochondria [36,45] (Table 1). SIRT6 and SIRT7 are nuclear, being present in heterochromatin and nucleoli, respectively [36]. However, during G1, but not the S phase of the cell cycle, SIRT6 is present in nucleoli, and when overexpressed it slows down mitosis [46]. More about the function of each in their respective locations follows.

Table 1 The activity and localization of each sirtuin. Sirtuin

Activity

Location

Targets

SIRT1

Deacetylation

Nucleus Cytosol

SIRT2 SIRT3

Deacetylation Deacetylation ADPRibosylation

Cytosol Mitochondria

SIRT4 SIRT5

ADP-ribosylation Deacetylation Demalonyation Desuccinylation ADP-ribosylation deacetylation

Mitochondria Mitochondria

FOXO1, FOXO3, PGC-1a, p53. NF-kB, Notch, HIF1a, LXR, FXR, SREBP1c, etc FOXO1, PEPCK, tubulin, PAR-3 OXPHOS complexes, SOD2, LCAD, HMGCS2, GDH, IDH2, PIP2, ACADL, FOXO3, ACSS2, OTC, GLUD1, NDUFA9, SDHA, ATP5A1, ALDH2, MRPL10, STK11, HISTH3, XRCC6 GDH CPS1

SIRT6 SIRT7

135

Nucleus Nucleolus

H3K9, H3K56

References to the information in this table can be found in the text and Ref. [45] in the case of mitochondrial sirtuins.

136

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

Enzymatic activity of each sirtuin Each sirtuin has a characteristic enzymatic activity [47–49] (Table 1). SIRTs 1, 2, 3, 5, and 6 have NAD þ -dependent deacetylase activity [16,50]. Whether SIRT7 is [51] or is not [36] a deacetylase [3] has now been clarified by recent evidence pointing to an important role for SIRT7 in deacetylation of acetylated histone H3K18 [52]. Deacetylation is the most prominent activity of SIRT1 [16] and SIRT2 [39]. In contrast, SIRT4 [53] and SIRT6 [54] possess mostly mono-ADP-ribosyl transferase activity. SIRT3 [55] and SIRT 6 [26,54,56] possess both activities. SIRT5 was originally reported to deacetylate CPS1 [57], but its demalonylation [58,59] and desuccinylation [58] actions on CPS1 and other proteins now seem more important. It has therefore been suggested that sirtuins should be redefined as ‘‘deacylases’’ rather than deacetylases [58]. A 25 amino acid sequence in the C-terminal domain of SIRT1 is essential for SIRT1 activity, serving as an ‘‘on’’ switch for the deacetylase core [60]. By itself the catalytic core has low catalytic activity, but regions in the N- and C-terminal potentiate catalytic activity 12- to 45-fold [61]. NAD þ levels increase during exercise, fasting, and calorie restriction [62,63]. During such ‘‘energy stress,’’ NAD þ may serve as a metabolic sensor leading to SIRT1 activation. While the 2-fold range of intracellular NAD þ fluctuations encompasses the Km of SIRT1, it is not clear how much NAD þ is actually bioavailable [63]. Conversion of NAD þ to its reduced form, NADH, lowers sirtuin activity [64]. A high-fat diet reduces the NAD þ /NADH ratio and sirtuin activity in mice [65]. NAD þ can be synthesized from tryptophan [66] and the dietary vitamin B3 components nicotinic acid and nicotinamide [67], as well as nicotinamide ribose present in milk [68]. Although the NAD þ precursor nicotinamide mononucleotide can activate SIRT1 and improve glucose tolerance, it is not present in the human diet [69]. Whereas at low levels nicotinamide serves as a NAD þ precursor, thus producing an increase in SIRT1 activity, at high levels nicotinamide exerts potent, end-product inhibition of SIRT1 in a manner that is noncompetitive with NAD þ [70,71], making nicotinamide deleterious to the cell [72]. Nicotinamide, by increasing the NAD þ /NADH ratio, induces SIRT1-mediated autophagy [73]. A conserved serine in the catalytic domain of SIRT1 can be phosphorylated in response to cAMP-mediated PKA activation, leading to increased SIRT1 activity independent of NAD þ [74]. This may have relevance to b-adrenoceptor- or cold-mediated stimulation of fatty acid oxidation and energy expenditure [74]. Alterations in the activity of enzymes that deplete intracellular NAD þ also affect sirtuin activity. The protein nimbrin, involved in DNA repair, is mutated in Nijmegen breakage syndrome, a condition exhibiting elevation in ROS and hyperactivation of PARP enzymes, which consume and thereby lower NAD þ , thus reducing SIRT1 activity [66]. But when PARPs are inhibited, SIRT1 is activated [75]. Since SIRT1 and PARPs are confined to the nucleus, PARP gene deletion has no effect on SIRT2 or SIRT3 [75,76]. CD38 also consumes NAD þ , and CD38 deletion increases sirtuin activity [77]. Thus sirtuins have unique and overlapping activities and their activation is exquisitely sensitive to modulation of NAD þ /NADH ratios.

Actions of nuclear sirtuins (SIRT1 and SIRT6) These are crucial in adaptation of metabolic processes to redox state. SIRT1 There is now considerable data on how dietary restriction exerts antiaging effects via SIRT1. Histone deacetylation was

noted first [78] and then later deacetylation of nuclear receptors and the transcriptional coactivator PGC-1a [79], FOXO proteins [80–82], and transcription factors and their cofactors (see reviews [34,49,63]). The activation of SIRT1 by calorie restriction stimulates PGC-1a to induce expression of gluconeogenic genes and reduces the repressive effect of PGC-1a on glycolytic genes, thus increasing hepatic glucose output [79]. By activating PPAR-a, SIRT1 increases fatty acid oxidation [83]. Screening for transcription factors that partner with SIRT1 in response to nutrient restriction identified HNF-1a, a homeobox protein involved in regulation of b-cell and hepatocyte function [84]. The HNF-1a–SIRT1 complex binds to two sites in the promoter of the gene for the well-known disease marker C-reactive protein. Deacetylation of FOXOs by SIRT1 [80,82] channels transcriptional regulation toward specific targets in pathways involved in stress resistance, lipid metabolism, and apoptosis [85]. Whereas deacetylation of FOXO3A by SIRT1 suppressed apoptosis genes, it activated stress resistance genes [80]. An additional stressderived signal is required to trigger the functional interaction of FOXOs and SIRT1 in the nucleus. PGC-1a is the master regulator of mitochondrial biogenesis, doing so by orchestration of a constellation of transcription factors needed for induction of gene expression in mitochondria [79,86]. Just as is the case for FOXOs, the coexistence of PGC-1a and SIRT1 in the nucleus does not lead to deacetylation unless additional signals concerning energy stress occur. This involves an imbalance in AMP/ATP ratio [87]. When this ratio is increased, SIRT1 and AMPK are activated [87]. The link between SIRT1 and AMPK is one of the most important connections to have emerged in recent years [64]. Each is activated under conditions of energy deprivation, such as occurs during calorie restriction. SIRT1 and AMPK work in synergy to adjust cellular physiology. AMPK phosphorylates, and thereby activates, PGC-1a [88]. This effect primes PGC-1a for deacetylation and activation by SIRT1 [64], although, in vitro, SIRT1 can deacetylate nonphosphorylated PGC-1a [89]. It has been suggested that the phosphorylation either modifies the nuclear localization or facilitates interaction with proteins capable of stabilizing the SIRT1–PGC-1a interaction [63]. FOXOs are also phosphorylated by AMPK in response to energy stress [90]. It was therefore suggested that a similar mechanism as for PGC-1a might explain why, despite their coexistence in the nucleus, the interaction of FOXOs with SIRT1 only occurs during energy stress. The AMPK/SIRT1/PGC-1a signaling pathway is utilized by several hormones, such as leptin, adiponectin, and fibroblast growth factor 21, to increase mitochondrial metabolism [91–93]. Hepatic effects of SIRT1 involve control of two pathways that affect gluconeogenesis in opposite ways. The activation by SIRT1 of FOXO1 and PGC-1a favors glucose production [79]. In contrast, deacetylation by SIRT1 of CRTC-2 leads to CRTC2 destabilization, with the result that glucose production is suppressed [94]. Each pathway is fine-tuned according to the length of fasting. Overall, with steady calorie restriction, the net effect is a mild increase in glucose output [64]. SIRT1 modulates carbohydrate metabolism by deacetylating other transcription factors. In the case of gluconeogenesis, which maintains blood glucose levels during fasting, regulation involves CREB, which in turn is controlled by CRTCs [95]. In the early stages of fasting CRTC2 translocates to the nucleus, where it activates CREB on promoters for genes such as PEPCK and G6P [94]. With prolonged fasting, increased NAD þ in the liver activates SIRT1, which deacetylates CRTC2, shunting it to degradation pathways [79,94]. In this way SIRT1, by attenuating gluconeogenesis, helps prevent premature energy depletion during the fasting state. The ability of SIRT1 orthologs to decrease CRTCs may be one mechanism by which abrogation of CRTC and CREB orthologs in C. elegans had been thought to prolong life span [96].

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

After deacetylation, both PGC-1a and FOXOs increase lipid catabolism and mitochondrial respiration [63]. SIRT1 also inhibits lipid anabolism. SIRT1 forms a complex with NCoR1 [97], leading to interference with PPAR-g signaling in white adipose tissue and LXR [98]. During fasting the suppression of PPAR-g activity results in fat mobilization instead of storage. The SIRT1–LXR pathway appears important for reverse cholesterol transport. The beneficial effects of SIRT1 on cholesterol metabolism may involve not only LXR deacetylation but also deacetylation of SREBP-1c. As a result, the affinity of SREBP-1c for promoters of lipogenic target genes is decreased and, via ubiquitination and ensuing degradation, SREBP-1c protein levels in the cell are reduced (see review, [63]). This could explain how SIRT1 might promote beneficial effects on cholesterol metabolism with no detrimental effect on lipid accumulation in the liver. In this regard SIRT1 prevents steosis during high-fat feeding [99,100]. The processes described above explain how SIRT1 is able to orchestrate metabolism at the cellular and whole-body level in order to extract energy from sources other than carbohydrate—in particular, pathways involving mitochondrial respiration (Fig. 1). By actions on circadian clock proteins [101], circadian rhythm appears connected to SIRT1’s deacetylation reactions that affect aging and metabolism. Feeding/fasting cycles influence the circadian clock via NAD þ levels, that then influence SIRT1 activity and thus metabolism [63]. SIRT1 assists in stress resistance via p53, HIF-1a, and HIF -2a, as well as HSF-1 [102]. It regulates the cellular response to oxidative and genotoxic stress by binding to eIF-2a, as well as effects involving the mediators of eIF2a dephosphorylation, growth arrest, and GADD34 protein and CreP [103]. SIRT1 is intimately linked to the formation of facultative and constitutive heterochromatin. This effect is crucial during the cellular response to stress. In the case of facultative heterochromatin formation, SIRT1 deacetylates the acetylated forms of histones H4K16, H3K9, and H1K26 [104]. It also promotes methylation of H3K9 in conjunction with the histone methyltransferase SUV39H1 [105]. SIRT1 is important for histone gene expression, which takes place during the S phase of the cell cycle. Histone gene regulation involves recruitment of SIRT1 and the CBP–p300 coactivator complex to histone promoters [106]. The role of SIRT1 in DNA repair involves actions on NBS1, PARP-1, Ku70, and WRN [102]. In the case of constitutive heterochromatin

137

formation, SIRT1 enhances SUV39H1 protein levels and turnover, thus leading to greater genomic integrity during the stress response [107]. There are also targets for SIRT1 in the cytosol. Cytosolic acetyl-CoA synthetase 1 is only deacetylated, and thereby activated, by SIRT1 [108], although the physiological role of this is unclear. SIRT1 activates eNOS in the cytoplasm [109]. This increases NO, leading to vasodilatation, increased blood flow, and nutrient delivery to tissues. Cytosolic SIRT1 regulates autophagy by deacetylation of key components of the autophagic machinery ATG5, ATG7, and ATG8 [110]. This prevents accumulation of damaged organelles, especially mitochondria, during starvation [110], consistent with SIRT1 being a master metabolic switch that drives the cell to derive energy from sources other than carbohydrate. SIRT1 represses NF-kB-dependent expression of STAT3, thus lowering mitochondrial respiration [111]. FOXO3A, by inhibiting c-MYC, represses a large number of nuclear-encoded mitochondrial genes, leading to reduction in mitochondrial copy number, mitochondrial proteins, respiratory complexes, and mitochondrial respiratory activity [112]. FOXO3A inhibits the increase in ROS and HIF-1a stabilization normally seen during hypoxia [112]. A hallmark of most diseases of aging is inflammation. Not surprisingly, SIRT1 is involved in countering inflammatory processes [63,64]. This involves negative regulation of NF-kB [113]. SIRT1 activity in cells is also altered by factors that regulate its transcription and thus its intracellular concentration, by posttranscriptional regulation and posttranslational modifications, as will be discussed later. SIRT6 Deficiency of this other nuclear sirtuin in mice leads to low IGF-1, severe hypoglycemia, and early death [32]. SIRT6-deficient cells show increased HIF-1a, glucose uptake, and glycolysis, and diminished mitochondrial respiration [56]. The high glucose uptake by muscle and brown adipose tissue leads to severe, potentially fatal hypoglycemia [56]. Loss of SIRT6 is accompanied by genome instability. This is because during oxidative stress SIRT6 is recruited to sites of DNA double-strand breaks, where it stimulates DNA repair through both nonhomologous end-joining and homologous recombination [114]. SIRT6 binds to, and mono-ADP-ribosylates,

Fig. 1. Proteins that are targeted by SIRT1 for deacetylation and the metabolic processes that these regulate. Protein targets in the nucleus that are implicated in transcriptional metabolic adaptations are shown on the right, and those in the cytoplasm are shown on the left. (Modified from [63]).

138

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

PARP-1, leading to increased PARP poly-ADP-ribosylase activity and DNA repair [114]. Overexpression of SIRT6 in ‘‘middle aged’’ and presenescent cells strongly stimulates PARP1-dependent homologous recombination repair, suggesting that in aging cells the precise homologous recombination pathway becomes repressed, giving way to a more error-prone nonhomologous DNA end-joining pathway [115]. Mice in which deletion of SIRT6 is neural specific are small at birth owing to low growth hormone and IGF-1, but exhibit catch-up growth and become obese in adulthood [116]. Moreover, histone H3 in the brain becomes hyperacetylated [116]. The occurrence of the latter in regions involved in neuroregulation points to a central role of SIRT6 in obesity prevention via modulation of neural chromatin structure. SIRT6 is a transcriptional regulator. A deficiency of SIRT6 alters expression of hundreds of genes [117–119]. Like SIRT1, SIRT6 is able to repress NF-kB activity [117]. In response to TNF-a, SIRT6 exhibits a highly dynamic response on mouse promoters across the genome, with more than half of SIRT6 target genes only becoming apparent in response to stress-induced signaling that involves NF-KB [118]. The RelA subunit of NF-kB recruits SIRT6 and is responsible for SIRT6 relocalization [118]. The epistatic interaction between RelA and SIRT6 determines the shaping of diverse temporal patterns of gene expression, including prominent genes involved in cell senescence and aging [118].

Actions of mitochondrial sirtuins (SIRT3, SIRT4, and SIRT5) These sirtuins help the cell adapt to reduced energy consumption [45]. SIRT3 SIRT3 is the major mitochondrial deacetylase [120]. It regulates the acetylation status, and thus activity, of metabolic enzymes such as acetyl CoA synthetase 2 [45]. During prolonged fasting, upregulation of SIRT3 in liver and brown adipose tissue leads to activation of long-chain acyl dehydrogenase and ornithine transcarbamoylase involved in b-oxidation of fatty acids and the urea cycle [121,122]. It could be that involvement of sirtuins in the production of ATP from catabolism of fat rather than carbohydrates may protect against ROS production and aging [123]. In support of this, SIRT3 deletion exacerbates diet-induced obesity [124]. Another target of SIRT3, HMGCS2 [125], regulates ketone body production, thus helping supply energy to the brain during fasting. During calorie restriction SIRT3 activates the TCA cycle components IDH2 [126,127] and GDH [120,126]. GDH then promotes metabolism of glutamate and glutamine to generate ATP, which results in insulin secretion. SIRT3 maintains basal ATP levels and regulates mitochondrial electron transport by stimulatory effects on mitochondrial respiration [128]. SIRT3 targets involved in OXPHOS were identified by a proteomic screen [129]. These included electron transport chain complex I components (NDUFA11 and NDUFS8) [128,129], complex II components (SDHA and SDHB) [129], complex III component (56kDa core I subunit) [130], and complex V components (ATP5A1, ATP5B1, and ATP5F1) [129]. Others targeted were proteins that bind to SIRT3 [129]. Thus SIRT3 controls the final stage of mitochondrial aerobic respiration and may be an important regulator of SDH activity in cells and mouse brown adipose tissue [129]. Despite these effects, SIRT3-deficient mice exhibit normal basal metabolic rate and adaptive thermogenesis [120]. SIRT3 protects cells from oxidative stress in several ways. One is by reducing ROS, since ROS production is elevated in SIRT3 knockout mice [130]. Another is SIRT’s ability to deacetylate and activate SOD2, a major mitochondrial antioxidant enzyme [131–133].

The activation of IDH referred to above [127] increases NADPH generation, leading to in an increase in the ratio of reduced-tooxidized glutathione in mitochondria and protection against oxidative stress-induced cell death via enhanced detoxification of ROS. SIRT3-dependent mitochondrial adaptations, by stimulating pathways associated with calorie restriction, might be a central component of age retardation in mammals [127]. Cardiac protection by SIRT3 involves activation of FOXO3A, MnSOD, and catalase, thus reducing ROS, a reduction in activation of Ras, and downstream signaling through the MAPK/ERK and PI3K/AKT (also known as PKB) pathways, blocking hypertrophy and interstitial fibrosis [134]. The ability of SIRT3 to reduce superoxide is accompanied by increased genomic stability [135]. Thus the SIRT3-mediated increase in efficiency of OXPHOS protects against the damaging effects of ROS. Other protective effects involving SIRT3 include prevention of cell death caused by hypoxia and staurosporine. SIRT3 does this by preventing loss of mitochondrial membrane potential, intracellular acidification, and the accumulation of ROS [136]. Senescence of human diploid fibroblasts induced by high glucose is reduced when the SIRT3–FOXO1 signaling pathway is activated [137]. SIRT3 also acts as a tumor suppressor [135]. This involves suppression of ROS. Consistent with this, SIRT3 is inactivated in many tumors [135,138]. When overexpressed in tumor cell lines SIRT3 reverses the Warburg effect [138], which is the metabolic reprogramming that occurs in cancer cells to enforce the production of ATP by glycolysis. SIRT3 appears to achieve this by reducing HIF1a, thus suppressing genes for glycolysis and angiogenesis [138]. Stimulation by resveratrol of the enzyme involved in the first step of adrenal steroid hormone synthesis, P450SCC, located in the mitochondrial inner membrane, involves SIRT3 and SIRT5 [139]. Interestingly, another enzyme required for cortisol synthesis, P45011b, is also stimulated by resveratrol [139]. SIRT4 This mitochondrial sirtuin appears to be involved primarily in metabolism. In pancreatic b-cells SIRT4, via ADP-ribosylation, represses GDH [53]. Thus SIRT4 and SIRT3 have opposing roles in GDH regulation [53,120,126]. They also have opposing roles in fatty acid oxidation [121,140]. What is not known is how SIRT3 and SIRT4 integrate similar nutrient states into opposite responses. The decline in SIRT4 in b-cells and liver during calorie restriction increases glutamine availability for catabolic metabolism, as well as glucose production in the liver. Depletion of SIRT4 in hepatocytes and myotubes, or in vivo, increases mitochondrial and fatty acid metabolizing enzymes, as well as SIRT1, which mediates the effect on fatty acid oxidation [140]. Thus inhibition of SIRT4 may have implications for type 2 diabetes, a condition in which ectopic lipid storage is elevated. SIRT5 The effects mediated by this mitochondrial sirtuin include activation of the first and rate-limiting enzyme in the urea cycle—CPS1 [57,141]. This facilitates the disposal of ammonia during calorie restriction, when amino acids are used as fuel sources. As stated earlier, the main enzymatic activity of SIRT5 is, however, demalonylation and desuccinylation [59], including of CPS1 [58].

Actions of the cytosolic sirtuin (SIRT2) SIRT2 deacetylates FOXO1 [142] and FOXO3 [143,144], thus implicating it in the diversity of processes that these key transcription factors regulate.

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

SIRT2 overexpression delays cell cycle progression [41]. It colocalizes with microtubles in the cytoplasm, where it deacetylates a-tubulin [39,145]. During mitosis SIRT2 increases [41]. It migrates transiently into the nucleus during G2/M transition [39], where it deacetylates histone H4 to modulate chromatin condensation during metaphase [43,146]. SIRT2 also deacetylates histone H3 [147]. These effects implicate SIRT2 in cell cycle regulation [146,148]. A specific target of SIRT2 is the acetyl transferase p300 [149]. Autoacetylation of p300 is a transcriptional regulatory checkpoint. Deacetylation by SIRT2 of a lysine in the catalytic domain of p300 restores binding of p300 to the preinitiation complex, thus affecting preinitiation complex assembly [149]. In turn, p300 is able to acetylate SIRT2 [150], thus attenuating its deacetylase activity and relieving the inhibitory effect of SIRT2 on p53 transcriptional activity [150]. SIRT2 binds to 14-3-3 b and g protein isoforms [151], which are involved in control of a multitude of signaling molecules such as kinases and phosphatases, as well as transmembrane receptors [152]. The SIRT2–14-3-3b/g interaction enhances the AKT-dependent deacetylation of p53 by SIRT2, thus reducing the effect of p53 on transcription [151]. The homeobox transcription factor HOXA10 is another SIRT2 binding partner [153], although more work is required to ascertain the biological role of this interaction. By deacetylating the p65 subunit of NF-kB, SIRT2 regulates the wide repertoire of NF-kB-dependent genes [154]. It stabilizes PEPCK, the rate-limiting enzyme for gluconeogenesis, by deacetylation [155]. SIRT2 affects Schwann cell myelination by deacetylation of the cell polarity protein PAR-3 [156].

Actions of the nucleolar sirtuin (SIRT7) SIRT7 associates with active ribosomal RNA genes and binds to histones and RNA polymerase I to stimulate transcription [157]. A proteomic analysis using HEK293 kidney cells identified 462 proteins bound by SIRT7, including 257 in the nucleus, 189 of which were nucleolar [158]. Its interaction with RNA polymerase I and upstream binding factor was confirmed, and new associations included 150 proteins involved in transcriptional processes involving both RNA polymerase I and II, as well as 32 associated with chromatin remodeling complexes, particularly members of the B-WICH complex known to associate with the RNA polymerase I machinery to facilitate rDNA transcription, and 30 relevant to proteosome/ubiquination processes [158]. By chromatin immunoprecipitation and sequencing, 276 binding sites for SIRT7 were identified in the genome, 241 of which were at proteincoding genes [52]. Most (74%) of the latter were close to the transcription start site where histone H3K18 binds and where H3K18 was shown to be a deacetylation target of SIRT7 [52]. Almost 60% of the SIRT7-binding sites contain the MAP kinasesignaling-dependent ETS transcription factor, ELK4 [52]. Mice with SIRT7 deletion develop inflammatory cardiomyopathy, cardiac hypertrophy, fibrosis, increased collagen III accumulation, hyperacetylation of p53, increased apoptosis, and reduced resistance to oxidative stress [51].

139

Fig. 2 shows transcription factors and targets in the SIRT1 promoter. Activators include FOXO1 [159], PPAR-a [161] and PPARb (also known as PPAR-d) [162], CREB [163], and the cell-cycle and apoptosis regulator E2F1 [164]. Repressors include PPARg [165], ChREBP [163], HIC1 [166] (via CTBP [167]), and PARP-2 (involved in DNA repair, apoptosis, and transcription) [76]. SIRT3 transcription is stimulated by ERRa which binds to the transcriptional coregulator PGC-1a, leading to changes in expression of genes for development and function of brown adipose tissue [168]. The outcome of the various transcriptional effects will be discussed in subsequent sections.

Posttranscriptional regulation The well-known posttranscriptional regulator HuR binds to the 30 UTR of mRNA for many different genes and enhances mRNA stability. HuR increases SIRT1 mRNA and protein levels [169]. Under oxidative stress the cell-cycle checkpoint kinase CHK2 phosphorylates HuR causing it to dissociate from SIRT1 mRNA causing HuR to decay [169]. Micro (mi) RNAs target specific sequences in the 30 UTR of mRNAs to cause mRNA decay [170]. Sixteen microRNAs can bind to the SIRT1 30 UTR [171]. The miRNAs miR-34a [172] and miR199a [173] destabilize SIRT1 mRNA. More on miRNA effects will be discussed later.

Posttranslational regulation Various posttranslational modifications of sirtuins 1 to 6 take place [174]. These direct sirtuins to specific targets. They can also increase activity, as occurs after phosphorylation of SIRT1 [175]. The pattern of SIRT1 phosphorylation by the cyclin B–CDK-1 complex affects the cell cycle and cell proliferation [175]. JNK1mediated phosphorylation during oxidative stress increases nuclear localization of SIRT1 and orients SIRT1 to substrates such as histone H3, although not p53 [176]. Curiously, in obesity and metabolic syndrome, while SIRT1 activity is reduced [160], JNK activity is elevated [177]. In contrast, deacetylation of p53 occurs when SIRT1 is phosphorylated at threonine 522 by DYRK-1 and DYRK-2 [178]. This prevents apoptosis during genotoxic stress,

Transcriptional regulation of sirtuin genes Most is known about SIRT1, whose expression is induced during low energy states such as nutrient or calorie deprivation [159], and repressed during energy excess, such as high-fat feeding [160].

Fig. 2. Transcriptional regulation of the SIRT1 gene. Shown is the SIRT1 promoter and several of the transcription factors that influence transcriptional activity by acting on either proximal or distal regions of the promoter. Once synthesized, the SIRT1 protein can enhance the activity of some positive regulators such as FOXOs, and inactivate repressor complexes such as p53, PPAR-g, and HIC1/CTBP. (Modified from [63]).

140

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

thus promoting cell survival [178]. Many other kinases phosphorylate SIRT1 at sites that differ between species. A ubiquitin-specific peptidase, USP22, stabilizes SIRT1 by removing polyubiquitin chains conjugated to it [179]. Since USP22 is one of the 11 death-from-cancer signature genes and this effect leads to suppression of p53, USP22 may have critical roles in embryonic development and cancer. SIRT2 phosphorylation by cyclin E–CDK2, cyclin A–CDK2, and p53–CDK5 inhibits SIRT2 activity [180]. SIRT2 is dephosphorylated by the dual specificity protein phosphatase CDC14B, which decreases SIRT2 abundance [41]. SIRT2 phosphorylation occurs late in G2, during mitosis, and into the period of cytokinesis [41]. CDC14B may provoke exit from mitosis when ubiquitination of SIRT2 results in its proteosomal degradation [41]. SUMO proteins can interact with (‘‘SUMOylate’’ ) human, but not mouse, SIRT1, thus increasing its activity [181]. This affects apoptosis, leading to speculation that SUMOylation serves as a switch between the survival and the death of a cell [181]. Desomoylation, which involves SENP protease, inactivates SIRT1, leading to cell death [181]. Posttranslational modifications may help specify particular substrates according to the circumstances. Moreover, SIRT1mediated deacetylation of PGC-1a requires phosphorylation of the latter by AMPK [63]. A considerable amount of work is, however, needed to decipher the various mechanisms by which SIRT1 specificity is regulated.

Formation of complexes with other proteins Sirtuin action is modulated SIRT1-binding proteins. Binding of the cell-cycle apoptosis regulator E2F1 inhibits SIRT1 activity [164]. This effect may regulate the induction of apoptosis in response to DNA damage. The protein AROS binds to the N-terminus of SIRT1, but not other sirtuins, to double its activity and inhibit p53 [182]. AROS reduction increases apoptosis [182]. In contrast, SENP-1 inactivates SIRT1 by binding to the C-terminus and desumoylating it, thus increasing p53 activity [181]. During fasting, NCoR1 and SMRT bind to SIRT1 and PPAR-g to downregulate adipogenesis mediated by PPAR-g [97] (Fig. 3A). In response to genotoxic stress DBC1 binds to the catalytic domain of SIRT1 and inhibits its activity [183,184] in mice on a high-fat diet, but not during fasting [185] (Fig. 3B). DBC1 loss leads to increased SIRT1 activity, SIRT1-mediated deacetylation of p53, and a reduction in p53-mediated apoptosis [183,184]. DBC1 gene knockout increases SIRT1 activity 2- to 4-fold in various tissues, causes p53 hypoacetylation, and protects against hepatic steosis and liver damage in mice on a high-fat diet [185]. This phenotype was noted in liver-specific SIRT1 knockout mice [83,186]. DBC1 was, moreover, able to disrupt a complex of SIRT1 with the histone methyltransferase SUV39H1, thus inactivating each enzyme [187]. The interaction of DBC1 with SIRT1 in response to DNA damage and oxidative stress requires ATMdependent phosphorylation of DBC1 at threonine 454, which creates a second binding site for SIRT1 [188]. DBC1 binds to ER-a and cooperates synergistically with the important coactivator of estrogeninduced gene expression and estrogen-dependent growth of breast cancer cells, CCAR1, to enhance ER-a function [189]. In contrast DBC1 reduces binding of SIRT1 to ER-a and thus lowers ER-a deacetylation [189]. Binding of SIRT1 to DBC1 disrupts the binding of DBC1 to CCAR1 [189]. In this reciprocal manner, SIRT1 and DBC1 exert major effects on ER-a activity. The histone methyltransferase LSD1 binds to the catalytic domain of SIRT1 resulting in convergent repression of Notch target genes (Fig. 3C). The latter involves SIRT1-mediated deacetylation of acetylated histones H4K16 and H1K26, and LSD1-mediated demethylation

Fig. 3. Proteins that form complexes with SIRT1 affecting its activity. (A). Binding of the NCoR1–SMRT complex to SIRT1 leads to blockade of the transcriptional effect of PPAR-g on PPAR-g target genes. (B). Genotoxic and metabolic stress caused by a high-fat diet results in DBC1 forming a complex with SIRT1, leading to SIRT1 inhibition. (C). LSD1 demethylates, while SIRT1 deacetylates, specific histone genes (H3K4, H4K16, and H1K26) leading to repression of transcription of genes targeted by Notch. Activation of the Notch pathway reverses this effect. (Adapted from [192]).

of H3K4 [190]. Notch, once activated, blocks the interaction of LSD1 with SIRT1 [190] (Fig. 3C). SIRT1 is a component of the PRC4 complex [191]. At least one of the other components, EZH2, is a histone methyltransferase, which, when overexpressed, upregulates all other PRC4 components [191]. These and other proteins that bind to SIRT1 should affect the ability of SIRT1 to deacetylate other SIRT1 substrates, including PGC-1a and FOXOs [63]. SIRT1, via its deacetylase activity, may also influence corepressor and coactivator complexes and thus the binding properties of these. In the case of SIRT7 a proteomic analysis referred to earlier has identified 462 proteins that bind to this sirtuin [158]. A major theme for many of these was regulation of transcription of RNA polymerase I. Roles for sirtuins in tissue functions Sirtuins have a multiplicity of different actions in most tissues in the body. Those involving SIRT1 are summarized in Fig. 4. Liver Fig. 5 depicts the action of each sirtuin in the regulation of hepatocyte function. SIRT1 activates gluconeogenesis and inhibits glycolysis [192]. Studies in vitro show that SIRT1 increases fat oxidation and downregulates the master regulator of fatty acid synthesis, SREBP-1c [100,193].

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

141

Fig. 4. Major tissues targeted by SIRT1 and key effects that result. (Adapted from [323]).

Fig. 5. The roles of the various sirtuins in glucose metabolism. Glucose taken up by the cell via the glucose transporter (GLUT) is catabolized by glycolysis into pyruvate, which enters the TCA cycle in mitochondria to generate energy. Under conditions in which energy supply is limited, glucose production by gluconeogenesis (dashed arrows) increases. This involves the conversion of pyruvate to oxaloacetate and subsequently to malate. In the cytoplasm malate is converted back to oxaloacetate, which is used by PEPCK to produce phosphoenolpyruvate. The latter is then available for conversion to glucose. In b-cells SIRT1 increases insulin secretion by repressing UCP2 transcription. In liver cells SIRT2 stimulates gluconeogenesis by deacetylating and increasing the stability of PEPCK. SIRT3 stimulates SOD2, thus decreasing ROS production, and also enhances cellular respiration by increasing the activities of complex I, complex II (via SDH), complex III, and IDH2. SIRT3 deacetylates GDH, stimulating its activity to affect gluconeogenesis and insulin secretion. By ADP-ribosylation SIRT4 inhibits GDH. (Modified from [192]).

142

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

Studies of liver-specific SIRT1-null mice show that the gluconeogenic G6K and PEPCK genes are normally downregulated by a signaling pathway in which SIRT1 positively regulates the gene for Rictor, a component of the mammalian TOR complex 2 (mTORC2), to trigger a cascade of phosphorylation of AKT and FOXO1 [194]. The hepatic overproduction of glucose, chronic hyperglycemia, and increased ROS production in SIRT1 null mice leads to oxidative stress-mediated impairment in mTORC2/AKT signaling in other insulin-sensitive organs, and consequent insulin resistance [194]. The latter is reversed by treatment with antioxidants. Knockdown of SIRT1 mRNA by delivery of adenoviral vectors containing SIRT1 short hairpin RNAs has no effect on hepatic triglyceride accumulation, but severely affects glucose homeostasis and gluconeogenic capacity [193]. This is not, however, seen in the liver-specific SIRT1 knockout mice above [194]. While there has been speculation that SIRT1 boosts gluconeogenesis by PGC-1a activation, leading to coactivation of CREB on the promoters of genes for gluconeogenesis [195,196], this was based on artificial overexpression of PGC-1a, whereas the evidence for such a mechanism during physiological modulation of PGC-1a is weak [197]. In fact, as evident from liver-specific SIRT1 knockout [194], under most circumstances SIRT1 activation in liver is associated with a reduction in gluconeogenesis [198]. SIRT1 binds to PXR which, apart from being a xenobioticsensing nuclear receptor with a major role in drug metabolism, is upregulated in liver during fasting and serves as a modulator of hepatic energy metabolism [199]. This SIRT1 interaction disrupts PXR binding to PGC-1a [199]. SIRT2 activates gluconeogenesis by deacetylation and increased stability of PEPCK [155] (Fig. 5). SIRT3 has several targets in hepatocytes [192] (Fig. 5). SIRT3 knockout mice respond to a high-fat diet by accelerated obesity, insulin resistance, hyperlipidemia, and hepatic steosis [124]. After a week on a high-fat diet SIRT3 expression is elevated, but by 13 weeks is decreased [124]. The lipogenic enzyme SCD1 was highly induced in these mice, and SCD1 deletion rescued the mice from liver steosis and insulin resistance [124]. During calorie restriction SIRT3 stimulates [120,126] and SIRT4 inhibits [53] GDH and thereby gluconeogenesis (Fig. 5). Less is known about the function of SIRT6, although in the Otsuka Long-Evans Tokushima Fatty rat, rosiglitazone ameliorates hepatic stenosis by activating SIRT6, AMPK, PGC-1a, FOXO1, LKB1, and adiponectin, thus reducing hepatic lipid accumulation [200]. Pancreas SIRT1 stimulates insulin secretion from b-cells [201,202] only in young mice [203]. This might be explained by the reduction in NAD þ that occurs with age [204]. In b-cells UCP2 affects the ability of glucose to modulate the ADP/ATP ratio and cause insulin secretion [205]. By reducing UCP2, SIRT1 permits better coupling and production of ATP in response to high glucose [202]. The ability of SIRT1 to stimulate insulin secretion involves other mechanisms further downstream [202]. SIRT1 levels in b-cells are modulated by miR-9, which reduces SIRT1 mRNA stability [206]. Glucagon-like peptide 1 lowers NAD þ /NADH ratio, thus reducing SIRT1 activity and binding to FOXO4, leading to increased FOXO4 activity [207]. FOXO4 activation stimulates bcell growth, thus increasing insulin secretion. These experiments show that SIRT1 is a negative regulator of b-cell proliferation. SIRT3 activates GDH [120,126], but an effect on insulin secretion is yet to be shown. Mitochondrial SIRT4 inhibits insulin secretion via suppression of GDH activity in b-cells, thereby downregulating insulin secretion in response to amino acids [53]. These effects are alleviated during calorie restriction. In insulinoma cells, SIRT4 is

overexpressed, leading to a decrease in insulin secretion in response to glucose [208]. Muscle SIRT1 regulates mitochondrial metabolism in cultured muscle cells [209]. Muscle-specific knockout SIRT1 mice show that SIRT1 is not needed for deacetylation of PGC-1a nor for mitochondrial biogenesis induced by exercise [210]. Acute exercise leads to increased nuclear SIRT1 activity, but not protein, accompanied by increased expression of genes activated by PGC-1a, as well as of mitochondrial biogenesis, possibly via AMPK activation [211]. SIRT1 overexpressing mice have a normal phenotype under basal conditions [212]. During calorie restriction SIRT1 enhances insulin sensitivity of skeletal muscle by deacetylation and inactivation of the transcription factor STAT3, leading to decreased expression of the p55a/p50a subunits of PI3K [213]. This results in more efficient PI3K signaling in response to insulin. SIRT1 also binds to the p85 adaptor subunit of PI3K to stimulate PI3K-mediated insulin signaling in muscle cells [214]. During exercise muscle temperature can rise to 40 1C. Mild heat stress induces mitochondrial biogenesis and this correlates with activation of the AMPK–SIRT1–PGC-1a pathway [215]. The muscle wasting induced by glucocorticoid administration can be prevented by resveratrol-mediated activation of SIRT1 [216]. Calorie restriction reduces age-associated muscle atrophy by lowering oxidative stress, even when CuZnSOD (SOD1) is absent [217]. SIRT3 and MnSOD are increased in these mice [218]. SIRT3 is highly expressed in slow oxidative type I soleus muscles compared to fast type II muscles, and exercise training increases SIRT3 expression, phosphorylation of CREB, and upregulates PGC1a in skeletal muscle [218]. While fasting and calorie restriction increase SIRT3 protein levels, a high-fat diet suppresses SIRT3 [218]. Germ-line SIRT3 knockout mice exhibit reduced phosphorylation of AMPK and CREB, as well as lower PGC-1a expression [218]. Mice with muscle-specific deletion of SIRT3 exhibit marked hyperacetylation of mitochondrial proteins, but do not have the high-fat diet-induced mitochondrial dysfunction nor the metabolic abnormalities seen in germ-line SIRT3 knockout mice [219]. In resting muscle SIRT3 is localized to subsarcolemmal and intermyofibrillar mitochondria and with chronic stimulation SIRT3 is upregulated in an AMPK-independent manner [220]. Adipose tissue Lipid metabolism and homeostasis are affected by actions of sirtuins on various tissues, particularly white adipose tissue, liver, and skeletal muscle [192]. Fig. 6 shows the sirtuin targets and mechanisms involved. In fat cells, SIRT1 inhibits adipogenesis and increases lipolysis by suppressing PPAR-g activity [97]. This involves the ability of SIRT1 to promote the assembly of a corepressor complex that includes NCoR1 and SMRT on the promoters of PPAR-g target genes, thus repressing their transcription (Fig. 3A) and opposing fat storage during calorie restriction and fasting [97]. SIRT2-mediated inhibition of adipogenesis and promotion of lipolysis involves deacetylation-mediated activation of FOXO1 during nutrient deprivation [40]. SIRT2 enhances the binding of FOXO1 to PPAR-g and represses the transcription of the PPAR-g gene [40,142]. Brain and nerves SIRT1 sensitizes neurons to oxidative damage but also has neuroprotective effects [221]. Deletion of SIRT1 in neurons does not affect development of the CNS, although because of a deficiency in growth hormone the mice are smaller [222]. While glucose tolerance is affected little in younger mice, defects

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

143

Fig. 6. The roles of the various sirtuins in lipid metabolism. Fatty acids are taken up by the cell by fatty acid transporters, and then transported to mitochondria, where their oxidation leads to ATP production. HMGCS2 breaks down fatty acids to generate ketone bodies. In the cytosol, lipogenesis (dashed arrows) leads to synthesis of fatty acids from malonyl CoA by fatty acid synthase, after which they are converted to triglycerides. When energy demand is high, triglycerides can be broken down to free fatty acids by lipolysis, mostly in fat tissue. These are then released into the bloodstream. By enhancing lipolysis through the inhibition of PPAR-g and by decreasing fatty acid synthesis through SREBP1c, SIRT1 reduces fatty acid storage. SIRT6 represses expression of genes involved in fatty acid synthesis. SIRT3 activates LCAD to stimulate boxidation. By activating HMGCS2, SIRT3 stimulates ketone body formation. SIRT3 stimulates SOD2 to decrease ROS. SIRT3 enhances cellular respiration by increasing the activities of complex I, complex II, complex III, and IDH2. SIRT4 appears to be a negative regulator of transcription of genes involved in fatty acid oxidation. (Modified from [192].).

became greater as the mice age [222]. SIRT1 and SIRT5 proteins increase in cerebral tissue of rats in response to calorie restriction, and their rise is accompanied by enhanced cognitive ability [223]. SIRT1 mediates anxiety level, which is probably an adaptive response to environmental changes affecting food availability [224]. The mechanism involves deacetyaltion of brain-specific helix-loop-helix transcription factor NHLH2, thus stimulating monoamine oxidase A and reducing serotonin in the brain [224]. The prevention of aging-related neurodegeneration and impaired neuronal plasticity, memory, and social behavior by calorie restriction involves forebrain CREB-1 [225]. CREB directly regulates SIRT1 transcription [163] and recruits SIRT1 to assist it in the regulation of PGC-1a and nNOS transcription [225]. Loss of such functions may explain accelerated brain aging during overnutrition and diabetes. SIRT1 binds to and stimulates promoter activity of the presenilin1 gene [226]. Resveratrol stimulates both SIRT1 and presenilin expression, as well as proliferation of neuronal stem cells in the hippocampus of rats [226]. This points to a role for SIRT1 signaling in hippocampal plasticity. By deacetylating the critical epigenetic regulator methyl-CpGbinding protein 2, SIRT1 controls binding of this protein to BDNF in the hippocampus [227]. Treadmill exercise improves various important parameters in the rat CNS, especially the hippocampus, and is associated with increased SIRT1, mitochondrial biogenesis, and AMPK activation [228]. The hypothalamus is crucial for metabolic actions of SIRT1 [229]. In arcuate neurons of the hypothalamus, necdin, by

formation of a stable ternary complex with SIRT1 and FOXO1, is crucial for FOXO1 deacetylation and for modulation of the hypothalamo–pituitary–thyroid axis [230]. SIRT2 is the most abundant sirtuin in the brain. It is expressed in virtually all brain cells, but is highest in cells that produce myelin, namely the oligodendrocytes [145,231]. SIRT2 is severely depleted from the myelin proteome in PLP knockout mice, in which PLP and DM20 (which arise from the same primary mRNA transcript by alternative splicing) are absent [232]. PLP regulates SIRT2 protein levels via the QKI-dependent pathway [231]. By deacetylation of a-tubulin, SIRT2 decelerates cell differentiation of oligodendrocyte precursor cells [145]. The transcription factor Nkx2.2, via HDAC-1, binds to the start codon of the SIRT2 gene, and suppresses SIRT2 transcription in oligodendroglial cells, thus enhancing cellular differentiation [233]. In myelin sheaths SIRT2 acts at sites of microtubule remodeling [234,235]. In postmitotic hippocampal neurons SIRT2 inhibits neurite outgrowth and growth cone collapse [180]. There are, however, no studies to date in support of a direct role of SIRT2 in neuronal differentiation [148]. But since SIRT2 can deacetylate FOXO3, which can regulate the neuronal stem cell pool by maintaining senescence [236], it is possible that SIRT2 might be involved in the latter process [148]. SIRT1 also deacetylates FOXO3A and thereby modulates differentiation of neuroblastoma cells [237]. Since p53 can regulate neuronal growth and p53 can be deacetylated by SIRT2 in vitro, a role for SIRT2 in p53-dependent neuronal function is possible [148].

144

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

SIRT2 transcripts undergo alternative splicing [238]. The fulllength SIRT2 protein (SIRT2.1) is only moderately expressed in the central nervous system, the levels being comparable to those in other tissues. SIRT2.2 concentrations are, however, much higher, both during postnatal development and in adulthood. In the aged brain, an age-dependent increase in SIRT2.3 was observed [238]. In peripheral nerves, elaboration of myelin by Schwann cells is dependent on SIRT2 [156]. SIRT2 deacetylates the master regulator of cell polarity, PAR-3, thus lowering activity of the polarity signaling component atypical protein kinase C during the myelination program [156]. This may be relevant to peripheral neuropathy in diabetes [156]. Moreover, stimulation of SIRT2 may help in the treatment of multiple sclerosis, a disease involving progressive loss of myelin [148]. Mitochondrial SIRT3 serves as a prosurvival factor by protecting neurons exposed to NMDA-induced excitotoxicity [239]. The latter involves PARP-1 mediated NAD þ depletion. The ensuing rise in ROS is responsible for the increase in SIRT3.

Sirtuins in metabolism Mice with whole-body overexpression of SIRT1 are leaner, more metabolically active, and glucose tolerant [240,241]. This phenotype resembles calorie restriction. The mice were moreover resistant to diet-induced hyperglycemia, metabolic syndrome, and fatty liver [242,243]. Although they were protected against diseases of aging such as metabolic syndrome and cancer, there was no significant increase in their life span [19]. The higher SIRT1 activity achieved by its overexpression confirmed that NAD þ is sufficiently abundant in cells not to be rate limiting for SIRT1 activity. Physiologically, an increase in SIRT1 activity occurs during nutrient deficiency and exercise [159]. This correlated with elevated expression of nuclear-encoded genes for mitochondrial proteins and higher energy expenditure [159,244]. In early studies nutrient deprivation was found to increase expression of FOXO3A in concert with SIRT1 promoter stimulation [159]. p53 is another transcription factor that normally represses SIRT1. During nutrient depletion FOXO3A is activated and binds to p53, thus extinguishing its inhibitory effect [159]. SIRT1 and p53 negatively regulate each other by a homeostatic loop involving deacetylation and by transcriptional effects. Deacetylation of FOXO3A by SIRT1 enhances FOXO3A action, and this includes FOXO3A-mediated stimulation of SIRT1 transcription. Whereas the effect of FOXO3A on the SIRT1 promoter is indirect (via binding of p53), FOXO1 binds to DNA target sites in the SIRT1 promoter to activate transcription directly [245] (Fig. 2). By their separate actions both FOXO3A and FOXO1 generate a feed-forward mechanism involving SIRT1 expression and activity. It is notable, moreover, that AMPK also activates FOXOs [90] and increases SIRT1 expression [246]. The protein HIC1 binds to SIRT1, forming a corepressor complex on the SIRT1 promoter [166]. This action of HIC1 is regulated by its binding to the energy and redox stress sensor CTBP [167]. Since the binding of CTBP to HIC1 is increased by NADH, when glycolysis is low NADH decreases, leading to destabilization of CTBP/HIC1/SIRT1 inhibitory complexes, with the result that SIRT1 transcription increases. The consequent elevation in SIRT1 results in metabolic adaptation favoring utilization of energy sources other than carbohydrate. Chronic inflammation represses SIRT1. This involves induction by interferon-g of CIITA, which, with HIC1, attaches to the SIRT1 promoter to repress SIRT1 expression [247]. In obese mice and humans SIRT1 is reduced [160,248]. Nutrient overload activates PPAR-g, which then binds to PPAR-g response elements in the SIRT1 promoter to repress SIRT1

transcription [161,165] (Fig. 2). PPAR-a and PPAR-b/d can also bind to PPAR-g response elements, although for PPAR-b/d at least, the elevation in SIRT1 expression that ensues involves instead binding to p21, which has a stimulatory binding site in the SIRT1 promoter [162] (Fig. 2). The effects of each on lipid metabolism differ. PPAR-g causes lipid anabolism and inhibits SIRT1 expression, whereas PPAR-a and PPAR-b/d are involved in fatty acid oxidation and increased SIRT1 expression. More research is needed to understand whether differential sensing of lipid species may direct pathways in one direction or the other. The NAD þ -dependent protein PARP-2 binds to the SIRT1 promoter, thus inhibiting transcription [76] (Fig. 2). PARP-2 increases PPAR-g transcription, but not that of PPAR-a and PPAR-b/d [249]. PARP-2 deletion has a similar effect on bodily processes as SIRT1 activation [76]. This includes protection against diet-induced obesity, elevation in mitochondrial number, enhanced oxidative metabolism, and insulin resistance. During fasting, glycogenesis and glycogenolysis in the liver are activated early, followed by ketogenesis when fasting is prolonged. The fasting hormones glucagon and norepinephrine increase cAMP signaling, which elevates CREB. Gluconeogenesis is activated by at least two transcriptional pathways [250] (Fig. 7). One involves glucagon-mediated dephosphorylation of CRTC2 in the cytoplasm, causing it to move to the nucleus and serve as coactivator for CREB [94]. At the same time glucagonmediated activation of cAMP-dependent PKA phosphorylates CREB (Fig. 7). With longer fasting (12–18 h in mice), SIRT1 deacetylates CRTC2, permitting ubiquitination and degradation of CRTC2, thus attenuating CREB [94]. During this period, SIRT1 activates FOXO1 and its coactivator PGC-1a [79,80], which maintains gluconeogenesis after the loss of CRTC2. The regulation of SIRT1 transcription during fasting involves a short promoter sequence containing overlapping sites for CREB and ChREBP (Fig. 2), known to be activated by feeding and which mediate energy disposal and storage [251]. ChREBP suppresses SIRT1 transcription, whereas CREB activates it [163]. During fasting ChREBP translocates to the cytosol, meaning that its binding site on the SIRT1 promoter becomes exposed. Binding of CREB within this composite response element then stimulates SIRT1 transcription [163]. This effect of CREB occurs by 18 h of fasting and coincides with activation of SIRT1 as a result of the elevation in NAD þ generated by gluconeogenesis. SIRT1 limits the early phase of gluconeogenesis by deacetylating CRTC2 and, by activating PGC-1a, promotes the onset of the prolonged phase of fasting. At this time deacetylation of SREBP1 represses SREBP1 activity, thus shutting down fat and cholesterol synthesis [99]. By 24 h of fasting SIRT1 transcription returns to normal, most likely as a result of CRTC2 destruction, and such normal SIRT1 transcription appears to be sufficient for ongoing gluconeogenesis during prolonged fasting (Fig. 7). Gluconeogenesis then needs to be switched off quickly when feeding resumes. This involves insulin. Upon refeeding, insulin signaling stimulates AKT, leading to phosphorylation of FOXO1, which causes FOXO1 to shuttle to the cytoplasm. During this time ChREBP moves to the nucleus, binds to a site in the SIRT1 promoter that overlaps the CREB site, and represses SIRT1 transcription [163] (Fig. 7). Thus when insulin is high and glucagon is low PGC-1a and CREB actions are turned off. The SIRT1 substrate PGC-1a activates not only gluconeogenesis but also biogenesis of mitochondria, and PPAR-a, another substrate activated by SIRT1, stimulates mitochondrial fatty acid oxidation [83]. Thus SIRT1 upregulation early in fasting would boost oxidative metabolism in cells, thus assisting in the use of fatty acids, amino acids, and acetate as energy sources involving the gluconeogenic pathway.

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

145

Fig. 7. Nutrient availability regulates SIRT1-mediated metabolic response. Short-term (or early phase of) fasting causes a decrease in blood glucose and insulin levels, and a concomitant rise in glucagon. Both of these hormonal changes initiate cellular pathways that induce gluconeogenesis. Low insulin signaling results in the dephosphorylation of the transcription factor FOXO1 and its translocation to the nucleus. Glucagon produces an increase in cyclic AMP levels and activation of PKA, which phosphorylates CREB and drives its translocation to the nucleus. Glucagon also causes the dephosphorylation of the CREB coactivator CRTC2 to trigger its translocation to the nucleus. The transcriptional complex CREB–CRTC2 activates the transcription of genes for gluconeogenesis and SIRT1. In a second phase of fasting, after more prolonged deprivation of food, increased SIRT1 deacetylates CRTC2 and targets it for ubiquitination and degradation by the proteasome, thereby terminating the transcriptional activity of CREB. SIRT1 also deacetylates PGC-1a, FOXO, and PPAR-a increasing their transcriptional activity. PGC-1a coactivates PPAR-a to induce the expression of fatty acid oxidation genes, and FOXO1 to maintain the expression of genes for gluconeogenesis. In the fed state, SIRT1 expression is repressed by the activity of the transcription factor ChREBP, which translocates to the nucleus.

Fasting also leads to upregulation of mitochondrial SIRT3 [121], which then deacetylates enzymes for oxidative metabolism and for detoxification of ROS [252]. SIRT3 knockout mice exhibit decreased oxygen consumption, accompanied by increased production of ROS and oxidative stress in skeletal muscle, leading to activation of JNK, along with impaired insulin signaling [130]. The microRNA miRNA-34a is elevated in diet-induced and genetically obese mice [253] and serves to negatively regulate SIRT1 expression at the posttranscriptional level by binding to the 30 UTR of SIRT1 mRNA [172,253].

Chemical compounds that activate sirtuins Given the health benefits of sirtuins, it makes sense that chemicals that activate sirtuin activity might prove useful in pharmacotherapy [3,35,254–256]. The natural polyphenol, resveratrol, has long been known to confer health benefits. The first demonstration of this was its ability to prevent skin cancer in mice [257]. Then a decade ago, screening an array of chemical compounds for the ability to activate SIRT1 and extend C. elegans life span led to the identification of not just resveratrol but also quercetin, piceatannol, and several other natural polyphenols with this ability [6]. In 2007, large-scale screening of thousands of unselected compounds identified several small molecular activators of SIRT1, including SRT1720, that were up to 1000 times more potent than resveratrol in their ability to activate SIRT1 and exert beneficial effects [258,259], including quenching of the attenuated life span of mice fed a high-fat diet [259]. As noted in the Introduction, however, the ability of resveratrol to increase

SIRT1 activity by a direct effect has been questioned [260–262]. Earlier claims of direct activation were shown to be assay artifacts emanating from the fluorophore attached to the peptide substrate used in the assay [261–263]. Although, by use of peptide substrates lacking any chemical modifications, direct binding and activation of SIRT1 were subsequently demonstrated, the sequence of amino acids surrounding the lysine 382 residue of the short polypeptide substrates related to p53 that were used did not match the native sequence [264]. This means there is no compelling evidence for direct SIRT1 activation physiologically. The current view is that SIRT1 activation may involve an upstream effect [265]. Resveratrol activates AMPK [265–269]. The metabolic effects of dietary resveratrol are, moreover, lost in AMPK knockout mice [270]. In addition, resveratrol inhibits mitochondrial ATP synthase activity [271] by binding to its g-subunit [272]. By interfering with mitochondrial respiration in this way, resveratrol causes an increase in the ratio of AMP to ATP, which then causes AMPK activation [273] (Fig. 8). Such upstream effects probably explain how the various chemicals activate SIRT1 [274]. Resveratrol competitively inhibits phosphodiesterases responsible for degradation of cAMP [275]. The rise in cAMP activates the cAMP-regulated guanine nucleotide exchange factor EPAC1, which in turn increases intracellular Ca2 þ concentration [275]. This then activates phospholipase C and ryanodine receptor Ca2 þ release channel, leading to activation of the calcium/calmodulin-dependent protein kinase 1 and hence AMPK [275]. The ensuing rise in NAD þ activates SIRT1. Inhibition of the phosphodiesterase PDE4 by rolipram confers metabolic benefits similar to resveratrol, i.e., prevention of diet-induced obesity, increased physical stamina, glucose tolerance, and improved mitochondrial function [275]. Rolipram is,

146

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

Fig. 8. The action of resveratrol leading to increased SIRT1 activity. Some consider that resveratrol or its metabolites act on mitochondria as a mild ‘‘poison.’’ This then reduces ATP synthesis. As a result, the ratio of ATP to AMP decreases. The increase in AMP activates AMP kinase, which in turn increases NAD þ , which activates SIRT1. This then leads to the various effects of SIRT1. (Modified from [63]).

however, far more potent than resveratrol at blocking phosphodiesterases [276]. Moreover, SIRT1 activation by the SRT compounds is claimed not to involve phosphodiesterase inhibition [276]. AMPK can phosphorylate PGC-1a directly, thus triggering downstream effects without a requirement for SIRT1 activation [88]. SIRT1 appears, however, to be required for activation of AMPK by lower doses of resveratrol [3]. At higher doses of resveratrol AMPK activation is independent of SIRT1. In cultured cells at least, there is ample evidence that many of the effects of resveratrol and SRT1720 depend on activation of SIRT1 [3]. SIRT1 and AMPK are mutually reinforcing and interdependent for their full effects [277]. But whereas AMPK activation occurs within minutes, activation of SIRT1 requires hours, consistent with the latter being a downstream event [63]. Whereas resveratrol can activate AMPK in tissue from SIRT1 knockout mice [267,270], it is unable to activate SIRT1 in the absence of functional AMPK [270,277]. Activation of AMPK leads to activation of fatty acid oxidation, which increases NAD þ and hence SIRT1 [64]. The rise in NAD þ is sustained by an increase in NAMPT expression, leading to increased NAD þ synthesis from nicotinamide via the NAD þ salvage pathway [278]. A randomized, double-blind cross-over trial in humans found that the beneficial effects of 30 days of resveratrol were achieved at 150 mg/day [279], which was an order of magnitude lower than needed in mice. Just as in other species, the effects seen were generally similar to calorie restriction, and included reduction in metabolic rate and sleeping, activation of AMPK in muscle, as well as stimulation of SIRT1 and PGC-1a levels, increased citrate synthase activity, and improved mitochondrial respiration on a fatty acid-derived substrate [279]. Circulating glucose, triglycerides, alanine-aminotransferase, markers of inflammation, and blood pressure were reduced, and HOMA markers, that estimate b-cell function and insulin sensitivity, were improved [279].

Analysis of the exometabolome of HepG2 cells has shown that resveratrol causes a metabolic switch from glucose and amino acid utilization to fat utilization for energy production, consistent with an effect via AMPK and SIRT1 activation [280]. The cell cycle was slowed in the S phase without inducing apoptosis. Resveratrol can modulate alternative splicing of some primary mRNA transcripts, but does so by a SIRT1-independent effect [281]. As discussed in the Introduction, the life span-extending effects of resveratrol seen in lower organisms [282] are not supported by more recent findings. Furthermore, while resveratrol prevented mice on a high-fat diet from dying prematurely [244,265], it was unable to significantly increase the life span of mice on a normal diet [283]. In mice [244], but not humans [279], resveratrol increased mitochondrial content. The ability of SRT1720 to prevent diet-induced obesity and diabetes [258,259,269], as well as fatty liver [284], seems, moreover, to be indirect, probably involving AMPK activation [269]. Another study also suggested that SRT1720 and resveratrol work by indirect effects, but found, however, that SRT1720 was lethal to mice fed a high-fat diet, and that while insulin was decreased, plasma glucose was not and mitochondrial capacity did not improve [261]. SRT1720 and SRT2183 can, moreover, decrease p53 acetylation in the absence of SIRT1, apparently by inhibiting the histone acetyltransferase p300 [285]. It should be obvious that the most direct way of activating all sirtuins, not just SIRT1, is by raising intracellular NAD þ levels. The main source of NAD þ is probably from salvage pathways of other metabolites of adenine nucleotides such as nicotinamide and nicotinamide riboside [66]. The enzyme NAMPT is rate limiting in conversion of nicotinamide to NMN, which is then converted to NAD þ by NMN adenylyltransferase [68,286] (Fig. 9). NMN administration to mice protects against age-related diabetes, in part through SIRT1 activation [69]. Although nicotinic acid and nicotinamide supplementation can raise intracellular NAD þ , albeit in a tissue-specific manner, an increase in sirtuin activity has yet to be described [63]. Niacin is composed of nicotinic acid and nicotinamide and is used to treat tryptophan deficiency as well as hypercholesterolemia. It would make sense that sirtuin activation might be involved in niacin’s beneficial effects, especially as niacin causes white adipose tissue to secrete adiponectin [287,288], which activates AMPK in muscle and liver [92]. The increase in fat deposition caused by adiponectin involves elevation in SIRT1, FOXO3A, and PGC-1a mRNA [289]. On a normal or high-fat diet SIRT1 overexpression increased adiponectin approximately 40% [242]. The nicotinamide riboseinduced increase in NAD þ in mammalian cells led to activation of SIRT1 and SIRT3, increased oxidative metabolism, and protection against high-fat diet-induced metabolic abnormalities [290]. As noted earlier, the presence of nicotinamide ribose in cow’s milk points to a potential dietary therapy [68]. Although stimulation of NAMPT increases NAD þ and SIRT1 activity [278,291–293], this could, at least in part, be a result of greater nicotinamide clearance [70]. A NAMPT inhibitor reduces NAD þ in the cytoplasm, but not in mitochondria, indicating that mitochondria lack the canonical NAD þ rescue pathway [294]. Another way to boost NAD þ availability for sirtuin activity could be via inhibition of other enzymes that utilize NAD þ , the major families being the PARPs and the cADP-ribose synthases CD38 and CD157 [66]. SIRT1 activity is downregulated during PARP-1 activation [75,295], and since SIRT1 is not directly affected by poly-ADPribosylation [75], it seems that PARP-1 and SIRT1 compete for a limited pool of NAD þ in the cell. Owing to the very low Km and high Vmax of PARP-1 [296], PARP may limit SIRT1 activity, but the opposite will not apply. Inhibition of PARP-1 leads to higher SIRT1 activity, which then elevates mitochondrial oxidative metabolism, suggesting

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

147

Fig. 9. The position of NAD þ at a crucial hub of pathways that affect SIRT1 activity and thus metabolic regulation. NAD þ is most likely rate-limiting for SIRT1 activity and therefore its actions on various pathways under divergent metabolic conditions. Activation of AMP kinase increases NAD þ biosynthesis through precursor (nicotinic acid [NA], nicotinamide riboside [NR], and nicotinamide mononucleotide [NMN]) supplementation, or through inhibition of other enzymes that consume NAD þ (e.g., PARPs or CD38), leading to an increase NAD þ levels. (Modified from [63]).

a means of protecting against metabolic diseases such as those caused by diet-induced obesity [75]. PARP-1 inhibition also increased mitochondrial gene expression [75]. Drugs that inhibit PARP-1 [297] could block the progression of cancer in part by boosting oxidative metabolism and opposing the Warburg effect referred to earlier (i.e., the reliance of cancer cells on anaerobic glycolysis even when oxygen is plentiful, an effect crucial for malignant transformation). By their ability to inhibit PARP-1, such drugs would, via increased NAD þ , raise SIRT1 activity, and by inhibiting PARP-2, would increase SIRT1 gene expression. The ability of sirtuin activators to induce a switch from glycolysis to oxidative metabolism [138,298] would similarly contribute to an antitumor action. Inhibition of PARPs leading to increased SIRT1 activity should mimic calorie restriction, and thus the beneficial effects of SIRT1 on healthy aging [299]. The elevated PARP activity in aged tissues, accompanied by a reduction in SIRT1, supports this premise [63,204]. So too does the fact that glucose homeostasis is impaired, age-related diseases are elevated, and life span is attenuated in mice expressing an extra copy of the human PARP-1 gene [300]. Only SIRT1 is activated in response to PARP inhibition, there being no effect on SIRT2 or SIRT3 [75]. This is probably because only SIRT1 is located in the nucleus and there are different pools of NAD þ specific for different intracellular compartments, with each pool being regulated independently [301]. A further NAD þ -consuming class of enzymes, the cADP-ribose synthases referred to above, are another group of molecules that compete more effectively than SIRT1 for intracellular NAD þ , CD38 having a very low Km for NAD þ [302]. Since approx. 100 molecules of NAD þ are required to yield one cADP ribose molecule [303], even low levels of CD38 are likely to have a profound effect on intracellular NAD þ [63]. Although CD38 is mainly extracellular, small quantities are also present inside the cell, including the nucleus, endoplasmic reticulum, sarcoplasmic reticulum, and mitochondria [304,305], where CD38 would compete with SIRT1 for available NAD þ [306]. In mice lacking CD38 the tissue level of NAD þ is elevated 10- to 20-fold [305] and CD38 in nuclear extracts is several fold higher [306]. The elevation in NAD þ in such mice was accompanied by a doubling of SIRT1 activity and consequent deacetylation of targets of SIRT1 [306]. Use of specific CD38 inhibitors [307] could also be relevant to treatment of diet-induced diabetes and other conditions [63]. Activation of SIRT1 changes the genome-wide transcription profile to resemble in many ways that induced by calorie restriction [308,309]. The potential for SIRT1-activating compounds (STACS) in treatment and prevention of common diseases of aging [47] was what prompted the extensive molecular screening,

irrespective of structure, for SIRT1 activators [258]. Thus STACs, by direct and/or indirect actions, should engender metabolic fitness. STACS are now undergoing clinical trials. The six other sirtuins are also likely to be valuable therapeutic targets. Their activation will likely involve chemicals that differ from those that lead to SIRT1 activation. But, as noted, since NAD þ activates all sirtuins, modulation of NAD þ levels seems a strategy worthy of exploration [63]. Since SIRT1 is critical for embryonic development, studying the role of SIRT1 on whole-animal physiology and biochemistry cannot be done in germ-line knockout animals. These complications were overcome by developing mouse strains in which whole-body SIRT1 deletion could be induced after the mice reach adulthood [310]. The usual increase in mitochondrial biogenesis and function, AMPK activation, and increased NAD þ in skeletal muscle in response to a moderate dose of resveratrol did not occur in these knockout mice. But at high doses resveratrol did activate AMPK. These experiments showed that dose is critical for AMPK activation. In mice lacking SIRT1 there was, however, no improvement in mitochondrial function irrespective of dose of resveratrol. Glucose homeostasis and liver function did not differ between SIRT1 knockout and wild-type mice in response to resveratrol. Whether this means SIRT1 has only a minimal role in liver or whether there is either residual SIRT1 in the knockout mice or compensation from extrahepatic influences is unclear. As noted, AMPK is an important mediator of resveratrol action [270,310]. It seems that at low doses resveratrol stimulates SIRT1 upstream of AMPK by deacetylation of the AMPK activator LKB1 [310] and, as discussed above, inhibition by resveratrol of PDE4, thus elevating cAMP and CamKK2 [275]. It was argued, however, that the latter mechanism might only occur with high doses of resveratrol [310]. Once activated, AMPK stimulates enzymes for NAD þ production and the rise in NAD þ increases SIRT1 activity [64,270]. Based on experiments with C2C12 myoblasts, it could be that at lower doses resveratrol stimulates SIRT1-dependent phosphorylation of AMPK, whereas higher doses activate AMPK by a mechanism that does not depend on SIRT1 [310]. Interestingly, at lower doses of resveratrol, NAD þ and ATP are increased by 12 h, but at higher doses (50 mM) each is reduced [310]. It may be that the higher doses inhibit mitochondrial respiration, thus lowering ATP, and that this then leads to AMPK activation, independently of SIRT1 [310]. Since PGC-1a is the ultimate beneficiary of the signaling pathways induced, it will be important to determine whether SIRT1-dependent deacetylation of PGC-1a occurs prior to, coincident with, or after AMPK activation [311]. In this regard, it is of interest that a specific AMPK agonist, AICAR, increases NAD þ , SIRT1 activity, and PGC-1a deacetylation [64]. Clearly SIRT1 is a mediator of the effects of resveratrol, increased PGC-

148

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

1a protein and activity being in large part what then lead to prevention of diseases involving mitochondrial dysfunction and aging [19,64,265,283]. But whether the rise in NAD þ is an adaptation to drive SIRT1 effects long term or to replenish NAD þ consumed by the initial increase in SIRT1 activity is another issue to be resolved [311]. In insulin target tissue, resveratrol decreases iNOS and NO production in skeletal muscle and white adipose tissue, but not liver, of endotoxin-treated mice [312]. This was also seen in each cell type in vitro and involved activation of AMPK, but not SIRT1 [312]. Other SIRT1 activators include oxazolo[4,5–b]pyridines and related heterocyclic analogs [313], as well as imidazo[1,2–b]thiazole derivatives [314]. Whereas there are chemicals with a wide range of core structures that inhibit the other sirtuins [256], sometimes selectively, as for SIRT2 [315] and SIRT3 [316], activators are less common. It is therefore of interest that placement of a benzyl group at N1 of the scaffold structure of 1,4-dihydropyridine conferred potent SIRT1, -2, and -3 activation [317]. SIRT3, AMPK, and mitochondrial biogenesis are activated by the natural stilbene viniferin [318]. Viniferin also attenuates the huntingtin-mediated loss of SIRT3 in neurons [318]. SIRT3 was, moreover, essential for the neuroprotective effect exerted by viniferin. Clinical trials of resveratrol and SRT compounds in patients with various conditions are in progress and the limited results to date highlight the need for care in relation to dose [3,319]. A small clinical study with resveratrol has provided promising results for obesity [279]. For treatment of cancer, inhibitors of SIRT1 and SIRT2 have been proposed [256], but there is a long way to go before the various chemicals available reach the clinical trials stage.

Obesity In obesity SIRT1 activity is low, consistent with a causative role. The mechanism of SIRT1’s effect could, in part, involve hypothalamic control of food intake [229]. Genetic deletion of SIRT1 in orexigenic Ag-RP neurons of the hypothalamus and administration of the SIRT1 inhibitor EX527 reduce food intake and body weight via the melanocortin pathway [320,321], whereas knockout of SIRT1 in anorexigenic POMC neurons of the hypothalamus leads to obesity [322]. Perhaps SIRT1 is required for homeostasis, for example, by suppressing NF-kB. Thus, by disrupting the normal biology of different cells, deletion of SIRT1 in an anorexigenic cell type would cause obesity, whereas deleting it in an orexigenic cell type would lead to loss of body weight [323]. Mice lacking SIRT1 in SF1 neurons exhibit reduced energy expenditure and hypersensitivity to diet-induced obesity [324]. The converse is true for mice overexpressing SIRT1. It has been pointed out that to be effective in prevention of weight gain, SIRT1-activating compounds must be found that target only SF1 and POMC neurons, avoiding AgRP-expressing neurons [229]. Doing so would also avoid psychotropic side effects typical of current anorectic antiobesity drugs [325]. Various hormones control appetite by hypothalamic effects. Impairments in the action of orexin-A and leptin have, moreover, been implicated in the causation of obesity in SIRT1-deficient mice [324]. Transgenic SIRT1 overexpression in the striatum and hippocampus upregulates adipogenic genes in white adipose tissue and increases fat accumulation [326]. The mice exhibit decreased energy expenditure and lower mitochondrial gene expression in muscle, accompanied by impaired lipid and glucose metabolism, motor function, and hypothalamic hormone axis [326].

SIRT1 in fat cells inhibits adipocyte differentiation and suppresses inflammation by targeting PPAR-g and NF-kB [327]. The latter two factors might be expected to be increased when SIRT1 is knocked out. However, SIRT1 null mice exhibit lower body weight, smaller adipocytes, reduced adipocyte differentiation, reduced extracellular matrix content, lower adiponectin and leptin, decreased capillary density, lower angiogenic factors (apart from VEGF), reduced macrophage infiltration and inflammatory cytokine expression in adipose tissue, and lower macrophage differentiation of embryonic fibroblasts [327]. Perhaps SIRT1 controls adipose tissue function via regulation of angiogenesis, which is also controlled by inflammation. SIRT1 suppresses transcription of genes for inflammation, thus reducing inflammation in adipose tissue [328]. Both glucose control and control of fat synthesis involve subtle effects. SIRT1 knockout was found to predispose fat-fed mice to weight gain, hepatic triglyceride accumulation with fasting, and hepatic steosis in some studies [83,185,186,329], but not in another, in which mice with liver-specific SIRT1 deletion gained less weight, had better glucose tolerance, and were protected against hepatic steosis [62]. The latter study found no specific changes in response to a regular diet and the mice responded normally to calorie restriction [62]. Hepatic steosis and lipid accumulation in heterozygous SIRT1 knockout mice fed regular chow worsened with age [186]. Extendin-4 decreases body weight, free fatty acids, triglycerides, and heptic steosis by stimulating SIRT1 expression and downstream effectors such as AMPK in hepatocytes in vivo [330]. Lipolysis is stimulated when SIRT1 activation results in deacetylation and thereby activation of FOXO1, which binds to the promoter of the rate-limiting lipase for lipolysis, ATGL [331]. SIRT2 is low and HIF-1a is high in visceral adipose tissue from obese subjects [332]. Activation of HIF-1a in visceral white adipocytes is critical for diet-induced obesity, glucose intolerance, insulin resistance, and cardiomyopathy. Suppression of SIRT2 transcription leads to diminished PGC-1a and genes for b-oxidation and mitochondrial function [332]. As referred to above, screening of a vast array of compounds identified the natural phytoalexin, resveratrol, as a potent SIRT1 activator [6]. Although resveratrol can extinguish virtually all of the adverse effects of a high-fat diet in ob/ob mice, a strain that lacks leptin, obesity persisted [244,265]. Nevertheless diabetes, cardiovascular disease, cancer, hepatic steosis, and premature death were prevented [244,265]. As well, a shift in muscle fiber composition meant the mice could run twice as far before exhaustion [244]. Whereas life span shortening is averted in obese mice, resveratrol did not significantly extend their life span [283]. The beneficial effects of resveratrol in obese mice have been replicated in healthy obese humans [279]. As noted earlier, the dose needed (2 mg/kg/day) was 10-fold lower than that required in obese mice. The plasma levels achieved were, however, similar. After 30 days of treatment the subjects had reduced metabolic rate, as well as increased mitochondrial OXPHOS and respiration in skeletal muscle, but not number of mitochondria. There were increases in SIRT1 and PGC-1a protein, as well as activation of AMPK in skeletal muscle. Lipid was reduced in liver but increased in muscle, and plasma glucose, triglycerides, alanine-aminotransferase, and markers of inflammation were all decreased. Insulin sensitivity improved and systolic blood pressure was reduced by 5 mm Hg. The metabolic changes observed were reflected in the changes in gene expression seen by genome-wide expression profiling, with 219 genes upregulated and 250 downregulated [279]. The pathways affected were similar to those seen in obese mice treated with resveratrol [244]. There were no adverse effects. a-Lipoic acid activates SIRT1 in mice [333]. This occurs upstream of AMPK activation and leads to lowering of plasma

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

lipids and intracellular triacylglycerol accumulation [333]. The beneficial effects of a-lipoic acid on mitochondrial function and oxidative stress in high-fat fed obese mice involve stimulation of SIRT3 as well as SIRT1, leading to increased deacetylation of FOXO3A and PGC-1b and a reduction in hepatic steosis [334]. The microRNA miR-132, by decreasing SIRT1, lowers deacetylation of the p65 subunit of NF-kB, thus activating the latter and inducing IL-8 and MCP-1 transcription [335]. The p65 subunit is also a target of SIRT2, which, by causing its deacetylation in the cytoplasm, is involved in the regulation of NF-kB-dependent genes as well [154]. miR34a increases with disease severity in hepatic steosis and suppresses SIRT1 expression [336]. Maternal obesity of gestation is accompanied by diminished SIRT3 expression and mitochondrial function [337]. The resultant reduction in fatty acid oxidation in liver likely contributes to the obesity and associated comorbidities of this condition.

Type 2 diabetes Sirtuins have important roles in diabetes. Transgenic overexpression of SIRT1 prevents diabetes in various mouse models of this condition, as well as diabetes that occurs during normal aging [240,242]. Similarly, chemical activation of SIRT1 has antidiabetic and other beneficial effects. SRT1720 is one chemical being examined in clinical trials. The key NAD þ intermediate NMN, in part via activation of SIRT1, ameliorates type 2 diabetes seen with aging and caused by a high-fat diet in mice [69]. In addition to actions on liver, muscle, and fat cells, diabetes can be ameliorated by effects of SIRT1 in the hypothalamus, which is a major regulator of feeding behavior and energy expenditure [212,229,322]. The ability of resveratrol to improve glucose homeostasis involves in large part a SIRT1dependent pathway in the arcuate nucleus of the mediobasal hypothalamus, which has a pivotal role in integrating responses to peripheral alterations in nutrients and hormones [338]. SIRT1 increases insulin sensitivity by repressing the tyrosine phosphatase PTP1B [242,244,258,265,339] and increasing insulin secretion by suppressing UCP2 [202]. The antidiabetic drug metformin activates SIRT1 and AMPK, accompanied by a reciprocal reduction in p53 protein, which becomes deacetylated at a SIRT1 target site [340]. In diabetic retinopathy, metformin suppresses the metabolic ‘‘memory’’ of hyperglycemia stress in small and large blood vessels [341]. SIRT1 is intimately involved in mediating metformin’s beneficial effect. Resveratrol, by activating AMPK, SIRT1, and NF-kB, reduces retinal inflammation in diabetes [342]. Advanced glycosylation end products derived from food have a prooxidant effect and their contribution to insulin resistance in type 2 diabetes may involve suppression of SIRT1 expression [343]. The microRNAs miR-34a and miR-93 are increased in liver in middle and old age [344]. SIRT1 in hepatocytes is downregulated posttranscriptionally by miR-34a [253,344], miR-93 [344], and miR-181a [344] via a target site in the 30 UTR of SIRT1 mRNA. miR-34a and miR-93 also regulate Sp1, a transcription factor that regulates SIRT1 expression [344]. Insulin-resistant liver cells and serum from diabetic patients exhibit elevated miR-181a [344]. Overexpression of miR-34a in pancreas inhibits insulin secretion [344] and overexpression of miR-181a causes insulin resistance in liver cells [345]. Thus inhibition of miR-34a and miR-181a could help in the treatment of insulin resistance and type 2 diabetes. The peptide obestatin, derived from ghrelin, prevents apoptosis in 3T3-L1 preadipocytes by stimulating PI3K/AKT and ERK12 signaling, leading to phosphorylation of AMPK, induction of adiponectin, suppression of leptin, and inhibition of lipolysis [346].

149

Obestatin stimulates SIRT1 expression, which mediates the stimulation by obestatin of glucose uptake [346]. In mice fed a high-fat diet, obestatin ameliorates metabolic dysfunction [346]. Dietary restriction, by increasing SIRT1 levels and reducing inflammation, ameliorates renal injuries in diabetic Wistar fa/fa rats [347]. SIRT1 mediates in part the protective effect of resveratrol against mitochondrial function in renal mesangial cells, thus reducing oxidative stress and providing a possible therapy for diabetic nephropathy [348]. Each of NAD þ , resveratrol, and rapamycin can oppose mesangial cell senescence induced by high glucose, and this effect involves SIRT1 [349]. Downregulation of SIRT1, followed by an increase in acetylated FOXO4 and expression of the proapoptosis gene Bcl2I11, leads to apoptosis of glomerular podocytes [350]. The authors of this study therefore advocated stimulation of SIRT1 for prevention of podocyte loss in diabetic nephropathy [350]. Podocyte injury and mitochondrial damage caused by aldosterone can be prevented by SIRT1 activation and this preventive effect involves upregulation of PGC-1a [351]. Fig. 10 shows several of the SIRT1 targets associated with type 2 diabetes and the other conditions that will be discussed in the following sections.

Cardiovascular disease By various means the actions of SIRT1 protect against cardiovascular disease processes [352,353]. Humans who restrict their caloric intake exhibit a very favorable cardiovascular risk profile [354]. Calorie restriction, by increasing SIRT1, leads to deacetylation and thereby activation of eNOS [355]. The ensuing rise in NO causes vasodilatation and vascular protection. This may contribute to the ability of resveratrol (150 mg/day for 30 days) to reduce systolic blood pressure by 5 mm Hg [279]. SIRT1 is involved in protection against atherosclerosis [356]. Long-term calorie restriction in Rhesus monkeys delays the onset of cardiovascular and other diseases of aging [23,24]. Transgenic overexpression [357] or resveratrol-mediated activation [358] of SIRT1 reduces plaque formation. Susceptibility to atherosclerotic lesion formation is reduced by SIRT1-mediated activation of eNOS in endothelial cells [109] and inhibition of proinflammatory NF-kB signaling [113]. Moreover, the ability of AMPK to phosphorylate eNOS is needed for SIRT1 to deacetylate eNOS and thus affect NO generation [359]. Thus eNOS seems to be an important factor mediating the beneficial effect of SIRT1 on the cardiovascular system. Interestingly, SIRT1 expression in both endothelial and smooth muscle cells of resistance-sized vessels in the brain seems important for endothelium-dependent relaxation [360]. The protective effect of SIRT1 against atherosclerosis involves a reduction in macrophage foam cell formation [361]. This effect of SIRT1 is due to suppression of NF-kB signaling, which results in a decrease in expression of LOX-1 [361]. In fact transgenic mice with endothelium-specific blockade of NF-kB signaling exhibit reductions in markers of oxidative stress, diet- and age-related insulin resistance, vascular senescence, metabolic deterioration, as well as improvement in locomotor activity, upregulation of mitochondrial sirtuins, and increased life span [362]. While loss of SIRT1 in vascular endothelial cells does not affect the vasculature during embryonic development, it impairs revascularization in response to ischemia [363]. This includes a reduction in endothelial cell branching and proliferation, a decrease in blood vessel density, and a defect in the ability to form vascular networks. Part of the effect involves loss of SIRT1-mediated deacetylation of FOXO1, where normally SIRT1 deacetylation of FOXO1 opposes its antiangiogenic action [363]. FOXO1, -3, and -4 are essential for keeping vascular endothelial cells quiescent [364]. SIRT1 also blocks Notch and thus the ability of Notch to

150

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

Fig. 10. The numerous SIRT1 targets and regulatory proteins that are involved in various diseases of aging. (Expanded and modified from [3]).

coordinate essential steps in blood vessel morphogenesis [365]. An alteration in the interaction of SIRT1 and Notch signaling may contribute to bicuspid aortic valves, present in over 1% of the population, and that are a risk factor for aortic dissection and aneurysm [366]. Vascular remodeling induced by hypertension appears to involve SIRT2, which deacetylates microtubules, leading to redistribution of endothelial cells in response to angiotensin II and mechanical stretch [367]. SIRT1 expression is enhanced by pulsatile flow-induced shear stress [359]. Resveratrol by activating SIRT1 (indirectly) activates the MAP kinase-5/MEF2-dependent signaling pathway, thus lead¨ ing to upregulation of the transcription factor Kruppel-like factor 2 that controls expression of flow-responsive genes [368]. Activation of SIRT1 signaling pathways by resveratrol also inhibits the oxidative stress-dependent pathology of endothelial cells in response to high shear stress and proinflammatory factors [369]. Part of the vascular protection provided by SIRT1 appears to involve inhibition of expression of the angiotensin II receptor AGTR1 [370]. Knockout of AGTR1 reduces oxidative organ damage and increases renal SIRT3 and NAMPT [371]. The reduction in ROS was in fact mediated by the elevation in SIRT3. The life span of these mice was increased by 26%. SIRT1 plays an important role in preserving cell viability during hypoxia. In response to hypoxia SIRT1 activity and expression increase [372]. This increase is HIF-dependent. SIRT1 activates HIF-2a and promotes degradation of PHD2 [173,373], which controls the various HIF isoforms that are transcription factors involved in orchestrating adaptive responses in expression of genes for cell survival, metabolism, and angiogenesis in response to alterations in oxygen tension [374,375]. In contrast, SIRT1 inhibits HIF-1a [298]. SIRT6, which, like SIRT1, is located in nuclear chromatin, is a corepressor of HIF-1a [56]. In mitochondria SIRT3 inhibits HIF-1a via effects on ROS [138]. SIRT1 prevents premature senescence induced in endothelial cells by oxidative stress [376]. This effect of SIRT1 is mediated by the major stress sensor p53 [376]. Other actions of SIRT1 that protect

endothelial cells from senescence involve deacetylation of LKB1, leading to degradation of the latter, thus preventing prolonged LKB1-induced AMPK signaling [377]. SIRT1 also suppresses p66SHC, an adaptor protein, which, when inactivated, protects mice from vascular disease associated with aging [378]. SIRT1 does this by binding to the promoter and lowering histone H3 acetylation, thus blocking transcription. During oxidative stress, binding of SIRT1 to FOXO3A shifts its effects on transcription from apoptosis to stress resistance [80]. In endothelial cells FOXO3A, via an interaction with PGC-1a, stimulates transcription of enzymes such as MnSOD that are involved in scavenging ROS [379]. SIRT1 activation provides neuroprotection during cerebral ischemia by inducing autophagy [380]. NAMPT, which is rate limiting in NAD þ production, causes an elevation in SIRT1 activity, which then affects TSC2–TOR–S6K1 signaling to induce the autophagic proteins LC3-II and beclin-1 [380]. Hyperhomocysteinemia, a major risk factor of cardiovascular disease, is associated with increased oxidative stress leading to endothelial cell dysfunction. A reduction in methylenetetrahydrofolate reductase leads to increased ROS generation, eNOS uncoupling, reduced NO, lower homocysteine, impaired differentiation and increased senescence of endothelial progenitor cells, decreased telomere length, and downregulation of SIRT1 expression [381]. miR-34a, by downregulating SIRT1 mRNA, induces senescence of endothelial cells [382] and EPC-mediated angiogenesis [383]. miR-34a’s effect on senescence also involves modulation of FOXO1 [383]. The therapeutic benefit of atorvastin on coronary artery disease may involve inhibition of miR-34a, leading to upregulation of SIRT1 in EPCs [384]. Lysosomal dysfunction, via cathepsin-induced proteolytic cleavage of SIRT1, appears to play a critical role in stress-induced premature senescence of EPCs [385]. Another means by which SIRT1 protects against atherosclerosis is by effects on lipid and glucose, as well as metabolism in liver. SIRT1 increases HDL cholesterol by activating LXR [386] and the nuclear bile acid receptor FXR [387]. LXR activation in liver upregulates ABCA1, which then increases the transport of reverse

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

cholesterol from peripheral tissues [386]. LXR also activates the SREBP1 gene in liver. SREBP1 is a transcription factor that is highly active in the fed state and works to promote expression of lipogenic and cholesterogenic genes, thus increasing synthesis of fat and cholesterol. SIRT1, by deacetylating SREBP1 and thus suppressing its activity, inhibits lipid and cholesterol synthesis [100]. FXR activation results in its dimerization with RXRa, leading to favorable outcomes on lipids and glucose metabolism [387]. In a curious finding, one group found a proatherogenic effect of SIRT1 overexpression in mice fed an atherogenic diet [388]. This involved deacetylation of CREB. The ability of SIRT1 to improve glucose metabolism and insulin resistance in type 2 diabetes is another means by which SIRT1 counteracts atherosclerosis. Thus in various ways SIRT1 potently protects blood vessels against inflammatory and procoagulant stress signals. Sirtuins also exert cardiac benefits. For example, overexpression of SIRT1 protects the heart against pathological cardiac hypertrophy [389]. In heart failure, genes regulating mitochondrial function are downregulated. Cardiac hypertrophy and failure involves in part binding of PPAR-a to SIRT1, thus recruiting SIRT1 to the estrogenrelated response element and repressing mitochondrial genes in a RXR-dependent manner, just as occurs during fasting [390]. Activation of SIRT1 by resveratrol improves cardiac contractility, as well as left ventricular function, in trauma-hemorrhage [391]. PGC-1a and ATP increase, and c-MYC, cytosolic cytochrome c, and plasma TNF-a decrease [391]. The protective effect of SIRT1 on the heart during ischemia–reperfusion injury involves eNOS, NF-kB, and stimulation of autophagy [392]. Overexpression of IGF-1 propeptide helps the heart recover from an infarct and this may involve JNK-1signaling pathway-mediated stimulation of SIRT1 expression [393]. Postinfarction heart failure is associated with reduced SIRT3 as well as lower mitochondrial respiratory control index and higher mitochondrial permeability transition pore opening [394]. SIRT3, via modulation of mitochondrial homeostasis and proteins that mediate energy metabolism and adaptation to mitochondrial redox stress, is able to impede cardiac hypertrophy [395]. Overexpression of SIRT6 can suppress angiotensin II-induced cardiomyocyte hypertrophy by binding to the p65 subunit of NFkB, thus blocking the effects of NF-kB on transcription [396]. The NAD þ regulating enzyme NMNAT2 is downregulated in cardiac hypertrophy, and NMNAT2 overexpression blocks angiotensin IIinduced cardiac hypertrophy in a SIRT6-dependent manner [397]. In the case of SIRT7, homozygous knockout leads to cardiac hypertrophy, inflammatory cardiomyopathy, and reduced life span [51]. The kidney is important for blood pressure regulation. The protective effects of calorie restriction on the kidney with aging in mice are mediated by SIRT1 and activation of FOXO3 [398]. SIRT1 protects the tubular epithelium by maintaining peroxisome number and function, upregulating catalase, and reducing ROS [399]. Renal medullary cells are particularly prone to metabolic stress owing to local hypoxia and hypertonicity. Here too SIRT1 has a protective function, in part via induction of COX2 expression [400]. Overexpression of SIRT1, or activation of SIRT1 with resveratrol, upregulates PGC-1a, leading to improvement in mitochondrial function and epithelial–mesenchymal transition in renal tubulointerstitial fibrosis [401]. In a mouse model of renal lipotoxicity, SIRT3 reverses ROS production and inflammation in proximal tubule cells [402].

Neurodegenerative diseases A functional deficit in sirtuin activity appears to be involved in a variety of neurological diseases that increase in frequency

151

during aging. The cognitive decline that occurs with aging may be countered by sirtuin activation. SIRT1 improves learning and memory in mice [403,404]. Its expression in the hippocampus is crucial to such actions. This involves effects on dendritic branching, branch length, complexity of neuronal dendritic arbors, ERK1/2 phosphorylation, and its ability to alter expression of hippocampal genes involved in synaptic function, lipid metabolism, and myelination [404]. SIRT1 also represses miR-134 that normally downregulates CREB and BDNF to impair synaptic plasticity [403]. Another microRNA, miR34a, regulates neuronal stem cell differentiation in part by regulation of SIRT1 expression, but also p53 [405]. In a positive feedback loop, p53 induces expression of miR-34a, which suppresses SIRT1, thus preventing SIRT1-mediated deacetylation of p53, and thereby increasing p53 activity [406]. The most common serious neurodegenerative disorder is Alzheimer’s disease. In a model of this condition SIRT1 activation prevented axonal degeneration [407] and neurodegeneration [408]. SIRT1 is upregulated in mouse models of Alzheimer’s disease produced by transgenic overexpression of p25 (a toxic coactivator of CDK5), as well as in cultured neurons subjected to neurotoxic insults [408]. This could possibly represent a defense mechanism. By inhibiting NF-kB signaling in microglia SIRT1 protects against b-amyloid toxicity [409]. The increase in tau protein aggregates (‘‘tangles’’) in Alzheimer’s disease can be reversed by SIRT1’s ability to deacetylate tau, thus destabilizing this protein and reducing tangles [410]. In a mouse model of Alzheimer’s disease, overexpression of SIRT1 in the brain reduces toxic b-amyloid generated by proteolytic cleavage of the amyloid precursor protein [411]. A major aspect of this effect involves stimulation of a-secretase gene expression via SIRT1-mediated deacetylation of RAR-b, the transcriptional activator of ADAM10 (the a-secretase gene) [411,412]. As a consequence the amyloid precursor is directed along an alternate pathway that does not lead to b-amyloid production. In mice overexpressing both bamyloid and SIRT1, less plaque is formed. In addition, cleavage of the membrane-bound Notch receptor by a-secretase leads to upregulation of genes for neurogenesis [413]. The amelioration of neuronal stress in the brain, and in cultured neurons, by SIRT1 involves suppression of the serine/threonine Rho kinase ROCK1, the action of which is to inhibit a-secretase and thereby amyloid precursor protein processing in brain [414]. SIRT1 also promotes growth and survival of neurons in the CNS by downregulating TOR [415]. Leptin activates AMPK and SIRT1 in neuronal cell lines; thus the association of low plasma leptin with increased risk of Alzheimer’s disease may involve a reduction in SIRT1 [416]. Leptin’s ability to reduce phosphorylation of tau and production of b-amyloid is, moreover, sensitive to inhibitors of AMPK and sirtuins [416]. Vascular lesions have been implicated in age-related brain pathology [417]. The e4 variant of apolipoprotein E is strongly associated with Alzheimer’s disease. Unlike APOE3, APOE4 is unable to prevent inflammation in pericytes and the consequent disruption of the blood–brain barrier leads to leakage of bloodborne molecules that are toxic to neurons [418]. The vascular protection afforded by sirtuins may add to its role in the interplay between the vascular and the nervous systems in Alzheimer’s and other neurodegenerative disorders. It therefore seems reasonable to suggest that SIRT1 activation in the brain might help prevent or impede the progression of Alzheimer’s disease [419]. Loss of neurons with age is also seen in amyotrophic lateral sclerosis (ALS). In human postmortem ALS samples, while SIRT1 and SIRT2 were reduced in white and gray matter of the primary motor cortex, in cortical layers 1–VI of the precentral gyrus and ventral/dorsal horn of the spinal cord SIRT1, -2, and -5 were increased [420]. SIRT1 is upregulated in a mouse model of ALS

152

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

involving transgenic overexpression of mutant SOD1 [408]. In this model the increase in SIRT1 occurs in cerebral cortical pyramidal cells, hippocampal pyramidal cells of area CA1–3 and dentate gyrus cells, thalamus, and spinal cord [421]. By activation of SIRT1, resveratrol protects against neurodegeneration in cell models of ALS [408,422]. In a mouse model of Huntington’s disease, overexpression of SIRT1 improves neuropathology, increases BDNF expression, and enhances survival, whereas brain-specific knockdown of SIRT1 exacerbates pathology [423,424]. SIRT1 activates TORC1 and promoted its interaction with CREB [423]. SIRT1 rescues Huntington’s disease neurons from the interference caused by mutant huntingtin protein on the TORC1–CREB interaction that represses BDNF transcription [423,424]. BDNF transcription was also a key effect of SIRT1 and TORC1 in normal neurons. Enhanced cell survival involved SIRT1-mediated FOXOA deacetylation [424]. Parkinson’s disease, dementia with Lewy bodies, and multiple system atrophy involve a pathogenic effect of the protein asynuclein. In models of Parkinson’s disease, resveratrol, by stimulating SIRT1 and autophagy, causes degradation of a-synuclein, further confirming the importance of SIRT1 in neuroprotection [425]. SIRT1 overexpression in brains of mice with an a-synuclein mutation increases life span [426]. This effect involves deacetylation of molecular chaperones HSF1 and HSFV, thus increasing the activity of each [426]. SIRT1 protects neurons against neurotoxicity induced by prion protein 106–126 [427]. By inducing autophagy, SIRT1 protects against the reduction in mitochondrial membrane potential caused by this prion. SIRT1 also protects against prion-induced Bax translocation to mitochondria and release of cytochrome c into the cytosol [427]. The beneficial effects of SIRT1 were in part mediated by ATG5 and the definitive marker of autophagy, LC3-II [427]. TDP-43 is a major pathological marker in an array of neurodegenerative diseases. TDP-43 forms a functional complex with FMRP and STAU-1 [428]. The complex binds to the 30 UTR of SIRT1 mRNA in neuronal cells to increase SIRT1 mRNA stability, and thus SIRT1 levels and cell survival [428]. Sirtuin agonists have potential as therapeutics for neurological diseases and neurological decline with aging. Such agents need to be able to cross the blood–brain barrier, however. While SIRT1 activation is neuroprotective, upregulation of the major sirtuin in brain, SIRT2, is toxic to neuronal cells. Chemical or siRNAmediated inhibition of SIRT2 rescued cells from toxicity induced by a-synuclein in a cell model of Parkinson’s disease [429]. In an animal model of Parkinson’s disease SIRT2 deacetylated FOXO3A, thus increasing Bim, which stimulates apoptosis [430]. This points to the use of SIRT2 inhibitors in the treatment of Parkinson’s disease. SIRT2 inhibition protects dopaminergic neurons against cell death in a Drosophila model and in a cell model of Parkinson’s disease [429]. Neuroprotection in response to SIRT2 inhibition is seen in a cell model and in a huntingtinchallenged Drosophila model of Huntington’s disease [431,432]. Inhibition of SIRT2 reduces sterol levels via a decrease in trafficking of SREBP-2 [432]. Manipulation of cholesterol biosynthesis at the transcriptional level mimicks SIRT2 inhibition, thus establishing that the metabolic effects of SIRT2 inhibition are sufficient to reduce mutant huntingtin toxicity [432]. In silico screening identified a brain-permeable SIRT2 inhibitor possessing these neuronal effects [433]. SIRT2 could have adverse effects in nerves by inhibiting lysosome-mediated autophagic turnover, increasing accumulation and aggregation of amyloid-b and other proteins, and thus making cells more vulnerable to the cytotoxic effects of such aggregates [434]. Thus SIRT2 could be a valuable therapeutic target for a range of neurodegenerative diseases.

Roles for the five other sirtuins in neurological function and disease will require further research.

Cancer There seems to be little doubt that sirtuins are involved in carcinogenesis. But owing to the complexity and diversity of their effects, the mechanisms remain elusive and there is as yet no consensus on precisely what the role of each is in cancer [435]. Overall, however, it would appear that SIRT2 and SIRT6 may be tumor suppressors, whereas, depending on the context, SIRT1 seems to be either a tumor suppressor or an oncogenic factor [435]. Does SIRT1 promote cancer? SIRT1 is overexpressed in several types of human tumors, and could serve to silence tumor suppressors, so promoting cancer development [436,437]. Increased SIRT1 expression has been reported in breast cancer [166,438], prostate cancer [439], colon cancer [440], hepatocellular carcinoma [441,442], pancreatic cancer [443], nonmelanoma skin cancers [444,445], acute myeloid leukemia [446], and in adult T-cell leukemia-lymphoma patients and cell lines [447], consistent with, but not necessarily proving, an oncogenic role. There are a multitude of possible mechanisms that might be involved. The basic reasons for SIRT1 overexpression in cancer are largely unknown [435]. Also not clear is whether SIRT1 overexpression causes or is a consequence of tumorigenesis [448]. SIRT1 overexpression is the most likely explanation for the loss of global acetylation of histone H4K16 and methylation of histone H4K20 seen in human tumors [449]. SIRT1 localizes to the promoters of tumor suppressor genes in which 50 CpG islands are hypermethylated [450]. This leads to their transcriptional silencing. Inhibition of SIRT1 in breast and colon cancer cells increases H4K16 and H3K9 acetylation at the promoters of tumor suppressor genes and their reexpression, in the absence of an effect on promoter methylation [450]. SRT1720 administration activates SIRT1 in breast cancer cells implanted subcutaneously into mice, leading to tumor cell migration and pulmonary metastasis [451]. A likely reason for the elevated SIRT1 expression in cancer is loss of a transcriptional repressor. One is HIC1. Mice lacking HIC1 are prone to cancer and exhibit SIRT1-dependent resistance to apoptosis in response to DNA damage [166]. Another negative regulator of SIRT1 expression is IKZF3 (also known as AILOS), loss of which leads to lymphoma accompanied by increased SIRT1 [452]. In various cancers, loss of p53 is seen. p53 binds to the SIRT1 proximal promoter (Fig. 2) to suppress SIRT1 expression [159]. So loss of p53 would increase SIRT1 expression. Also, SIRT1 might cause cancer by its ability to deacetylate p53 at one lysine residue, inhibiting p53 activity on the promoter of p21, and blocking p53-dependent apoptosis [50,453]. SIRT1 inhibition reduces tumor growth and inhibition of p53 seems to be a common theme in mediating this effect [454]. For example, SIRT1 inhibition in chronic myelogenous leukemia (CML) reduces growth of quiescent leukemia stem cells both in vitro and in vivo in a p53-dependent manner [455]. The Philadelphia translocation involving formation of a fusion gene of part of the BCR gene and the ABL1 gene is a cause of CML. BCR-ABL kinase is constitutively active and, via STAT5 signaling, activates SIRT1 in hemopoietic progenitor cells, thus promoting CML cell survival and proliferation, accompanied by deacetylation of SIRT1 substrates such as FOXO1, p53, and KU70 [456]. Inhibition of BCRABL partially reduces SIRT1 expression, and knockout of SIRT1

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

prevents BCR-ABL-mediated transformation of mouse bone marrow cells and CML-like disease, as does SIRT1 inhibition [456]. Interestingly, SIRT1 is dispensable for maintenance of the hemopoietic stem cell compartment in mice [457]. Casein kinase-2 inhibitors, by downregulating SIRT1, sensitize glioma cells to TNFa-induced apoptosis [458]. An alternatively spliced form of SIRT1 lacking exon 8, and as a result its deacetylase activity, is expressed in human and mouse tissues [459,460]. The splice variant retains its ability to bind p53 [460]. The alternative splicing of SIRT1 that generates this version is, moreover, regulated by p53 [459]. The SIRT1 splice variant is overexpressed in various cancers lacking wild-type p53, and has been suggested as a possible therapeutic target [460]. As well as low deacetylase activity, the alternatively spliced SIRT1 is sensitive to stress, and differs from SIRT1 in its protein and RNA stability, as well as its interaction with other proteins [459]. Research is needed to determine the role of the alternatively spliced SIRT1 in cancer. Studies in vivo have, however, suggested that SIRT1 does not affect p53-dependent functions [461]. Nevertheless, p53 can be downregulated by other sirtuins, namely SIRT2 [462] and SIRT3 [463]. Another effect of SIRT1 is deacetylation of the tumor suppressor protein Rb, reducing its activity, and thus stimulating the cell cycle [464]. In diffuse large-cell lymphoma and Burkitt lymphoma SIRT1 binds to and deacetylates the oncogene BCL6, so activating it [465]. Cambiol inhibits SIRT1 and SIRT2, leading to hyperacetylation and inactivation of BCL6 [465]. Since cambiol is well tolerated, it may be suitable for therapy. The protein EVI1 causes proliferation and abnormal differentiation of hemopoietic stem cells [466]. In cell lines and chronic myeloid leukemia patient samples containing EVI1, SIRT1 is upregulated and this involves the binding of EVI1 protein to a site 1 kb upstream of the SIRT1 gene [467]. Moreover, SIRT1, by binding to and deacetylating EVI1, can negatively regulate EVI1 activity [467]. Thus SIRT1 may have a role in EVI1-positive neoplasms. The oncogene c-MYC can bind to the SIRT1 promoter to induce SIRT1 expression [468]. SIRT1 then deacetylates c-MYC to reduce c-MYC activity, target gene expression, and cellular transformation [468]. It was suggested that this mechanism could block tumor initiation in premalignant cells. The pathogenesis of neuroblastoma involves N-MYC, which stimulates SIRT1 transcription, followed by binding of SIRT1 to N-MYC, which serves as a transcriptional repressor complex on the MKP3 promoter [469]. The importance of SIRT1 in tumorigenesis of neuroblastoma was demonstrated by inhibition of SIRT1 with cambiol [469]. It was suggested that cambiol could serve in prevention and treatment of this cancer. In brain cancer, neuroinflammation causes breakdown of the blood–brain barrier. While resveratrol reduces markers of neuroinflammation (MMPs and COX-2) in human brain microvascular endothelial cells, SIRT1 appears not to be involved [470]. Expression of c-MYC is elevated in colorectal cancer and, by inducing NAMPT expression, raises NAD þ and thus SIRT1 activity [471]. Moreover, c-MYC, by sequestering the SIRT1-binding protein DBC1 from SIRT1, increases SIRT1 protein [471]. Human fibroblasts immortalized with c-MYC undergo senescence and apoptosis in response to downregulation of SIRT1 [471]. SIRT1 binds to c-MYC and deacetylates it, thus increasing c-MYC stability [471]. SIRT1 also stimulates c-MYC gene expression [471]. Thus, in the context of deregulated tumors, this positive feedback loop may contribute to the development and maintenance of tumors. SIRT1, by triggering c-MYC transcription, increases the recruitment of c-MYC to the hTERT promoter, leading to enhanced life span of HUC-F2 fibroblasts [472]. In colorectal cancer a reduction in SIRT1 expression is

153

associated with changes in clock gene expression, with lower PER3 and higher ARNTL1, CLOCK, PER1, PER2, CRY1, TIPIN, and CSNKIE [473]. Induction of SIRT1 leads to different changes in PER1 in different colorectal cancer cell lines [473]. Expression of both DBC1 and SIRT1 is associated with poor prognosis of gastric cancer [474] and breast cancer [438,475]. The negative regulation of SIRT1 by DBC1 may retard tumorigenesis, suggesting that the balance in expression of each may have prognostic significance [475]. High SIRT1 expression in gastric carcinoma correlates with lymphatic metastasis [476]. Multidrug resistance in gastric cancer involves binding of ATF4 to the SIRT1 promoter to activate SIRT1 expression [477]. In hepatocellular carcinoma SIRT1 promotes tumorigenesis by activating the PTEN/PI3K/AKT signaling pathway, leading to mitotic entry, cell growth, and caspase-3 and caspase-7-mediated inhibition of apoptosis [478]. SIRT1 regulates DNMT1, a key enzyme responsible for DNA methylation [479]. Depending on which lysines are deacetylated, DNMT1 activity can increase or decrease. Complete deacetylation impairs cell cycle G2/M transition. SIRT1 reduces the ability of DNMT1 to silence genes for the tumor suppressors ER-a and cadherin-1 [479]. Oxidative damage causes SIRT1 to relocalize to CpG islands in promoters [480]. This could explain aberrant DNA methylation and transcriptional silencing in specific cancers. In ovarian cancer, FOXL2 downregulation or mutation is seen in granulosa tumor cells [481]. FOXL2 reduction is pivotal to tumorigenesis, ovarian aging, and premature ovarian failure [481]. SIRT1 suppresses FOXL2 activity on targets involved in the cell cycle and in DNA repair, so that inhibition of SIRT1 by nicotinamide limits proliferation by increasing FOXL2 [481]. As discussed earlier, more than 16 microRNAs modulate SIRT1 mRNA stability by binding to its 30 UTR [171]. The first to be identified as a SIRT1 regulator, miR-34a [172], serves as a tumor suppressor gene in neuroblastoma [482]. Expression of miR-34a is reduced in drug-resistant DLD-1 colon cancer cells, and introduction of miR-34a induces apoptosis in colon cancer cells by downregulating SIRT1 [172,483]. This miRNA also downregulates SIRT1 in pancreatic, prostate, brain, and liver cancers [171]. p53 induces miR-43a expression and this causes SIRT1 downregulation [171]. Thus mutation of p53 leads to increased SIRT1 expression. In human fibrosarcoma HT1080 cells, miR-520c and miR-373 suppress SIRT1 and TOR mRNA translation by targeting the 30 UTR of each [484]. This results in activation of the Ras/Raf/ MEK/ERK signaling pathway and NF-kB, leading to increased MMP9 and thus elevation in cell migration and growth [484]. SIRT1 upregulation in breast cancer samples is associated with a reduction in miR-200a, another microRNA that targets the 30 UTR of SIRT1 mRNA [485]. In oligodendrogliomas and glioblastomas expression of 26 microRNAs differs from control brain tissue and 7 of these could discriminate each type of tumor [486]. SIRT1 was one of the miRNA targets differentially expressed in gliomas [486]. Patients with leukemia exhibit aberrant histone deacetylase activity. Treatment of leukemia cells with inhibitors of histone deacetylase causes upregulation of the proapoptotic BCl2 family member Bax, whose translocation to mitochondria is normally prevented by SIRT1 [487]. Since NAD þ -independent histone deacetylase activity and sirtuins cooperate in leukemia cells to avoid apoptosis, a combination of sirtuin inhibitors such as sirtinol, cambiol, and EX527 with histone deacetylase inhibitors leads to synergy in exerting an antileukemic effect [487]. In PC3 and DU145 prostate cancer cells, studies with sirtinol have shown that SIRT1 upregulation promotes cell growth and chemoresistance [488]. Moreover, studies involving sirtinol treatment of LNCaP prostate cancer cells reveal pleiotropic effects that involve pathways other than merely sirtuin inhibition [489].

154

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

SIRT1 deacetylates cortactin, a cell motility stimulating protein overexpressed in various cancers. Inhibition of SIRT1 or cortactin expression attenuates migration and invasion of DU145 prostate cancer cells [490]. By inhibiting cell proliferation and promoting apoptosis, FOXO3 functions as a tumor suppressor [364]. Deacetylation of FOXO3 by SIRT1 and SIRT2 causes the E3 ubiquitin ligase subunit SKP2 to bind to FOXO3, thus promoting its ubiquination and degradation [144]. Downregulation of FOXO3 in malignant PC3 and DU145 prostate cancer cells is a result of elevation of both SKP2 and SIRT1 [144]. Thus loss of FOXO3 caused by SIRT1 and SIRT2 upregulation might be considered pathogenic in some cancers. Inhibition of SIRT1 activity by a mutant version of the 25 amino acid C-terminal SIRT1 activator sequence, via its binding to the deacetylase core of SIRT1, increases the chemosensitivity of androgen-refractory prostate cancer cells [60]. This is consistent with a role for SIRT1 in supporting such prostate cancers. Both NAMPT, which generates NAD þ , and SIRT1 were overexpressed in prostate cancer cells, accompanied by increases in FOXO3A, catalase, and MnSOD, consistent with a role in carcinogenesis [491]. Interestingly, rather than being localized mostly in the nucleus, in cancer cells SIRT1 is located primarily in the cytoplasm [492]. This marked location aberration in cancer cells results from an elevation in stability of SIRT1 protein as a result of PI3K/IGF-1 receptor signaling. SIRT1 was, moreover, shown to be required for PI3K-mediated cancer cell growth [492]. The cytoplasmic location of SIRT1 was said to greatly contribute to its cancer-specific function.

inhibits apoptosis, resveratrol, by activating SIRT1, reduces survivin and thereby increases apoptosis [495]. Although SIRT1 might appear to be causative in cancer by inhibiting p53-dependent apoptosis [50,453], it may be that the other posttranslational modifications that p53 undergoes determine its specific activity. This has led to the concept that a ‘‘complex barcode’’ underlies p53’s actions [501], and to the suggestion that the term p53 ‘‘specification’’ rather than ‘‘activation’’ should be used [63]. In support of this Canto´ and Auwerx have used as an example the effect of p53 on mitochondrial metabolism. They pointed out that activation of p53 increases mitochondrial oxidation, whereas deacetylated p53 is associated with defective mitochondrial respiration. Yet, in the context of DNA damage, activated p53 decreases mitochondrial biogenesis [502]. Thus biological context and ‘‘barcode’’ of posttranscriptional regulation could be important determinants of the effect of p53, and no doubt other transcription factors. The ability of SIRT1 to suppress tumors seems to include deacetylation and inactivation of HIF-1a and thus genes targeted by the latter [298]. It has been speculated that if SIRT1 elevation in many cancers is a consequence, rather than a cause, of cancer then, given the ability of SIRT1 to inhibit senescence and apoptosis, it might be that some tumors could become ‘‘addicted’’ to SIRT1 [435]. Anticancer drugs and SIRT1 HBX protein, by binding to SIRT1, attenuates SIRT1 binding to

b-catenin, thereby upregulating b-catenin [503]. Stimulation of An anticancer role for SIRT1? There is also a body of in vivo evidence pointing to a tumor suppressor role for SIRT1 [19,436,492,493]. SIRT1 suppresses growth of intestinal and colon cancers [494], liver cancer [19], ovarian cancer, glioblastoma, and bladder cancer [436]. SIRT1 has a protective effect against breast cancer in mouse models [495] and colon cancer in human tumor specimens [494]. The latter involves an ability of SIRT1 to move an oncogenic form of bcatenin from the nucleus to the cytoplasm, limiting b-catenin signaling in colon cancer [494]. In response to SIRT1 activation, proliferation of pancreatic cancer cells is inhibited by downregulation of b-catenin and its target molecule cyclin-D1 [496]. In El4 lymphoma cells, resveratrol upregulates SIRT1 and downregulates NF-kB, thus promoting apoptosis, and having the potential to suppress tumor growth [497]. Resveratrol at 50 mM, but not 25 mM, inhibits SIRT1 expression and activity in Hodgkin lymphoma L-428 cells [498]. Both doses increase p53. These findings support the anticancer effect of resveratrol. Another small molecule, Inauhzin, by inhibiting SIRT activity, promotes p53-dependent apoptosis of lung cancer cells without exerting genotoxic stress [499]. Mice transgenic for SIRT1 exhibit a global decrease in carcinomas and sarcomas, but no alteration in lymphoma incidence [19]. These mice had less DNA damage and a reduction in p16INK4A. Calorie restriction is well known to reduce cancer incidence, yet in p53-deficient mice engineered to have 2- to 3fold overexpression of SIRT1, the increased cancer protection and life span extension that calorie restriction conferred were no different than in p53-deficient mice lacking SIRT1 overexpression, suggesting only a limited role for SIRT1 in cancer protection in response to calorie restriction [500]. In breast cancers associated with mutant BRCA1, SIRT1 is low and the apoptosis inhibitor survivin is high [495]. The wild-type form of BRAC1 binds to the SIRT1 promoter to increase SIRT1 expression, and SIRT1 then suppresses survivin. Since survivin

SIRT1 should therefore avoid resistance to anticancer drugs in HBX-related hepatocellular carcinoma [503]. The cancer drug doxorubicin causes genotoxic stress in both cancer cells and noncancer cells. By inhibiting AMPK it caused SIRT1 dysfunction and accumulation of p53 [504]. Thus pharmacological activation of AMPK and SIRT1 by resveratrol should lower the side effects of doxorubicin [504]. A side effect of doxorubicin is cardiovascular dysfunction. This involves a PARP2-mediated reduction in SIRT1 in blood vessels [505]. Inhibition of SIRT1 increases the sensitivity of breast tumor cell lines to the chemotherapeutic agent paclitaxel, doing so via activation of caspase-2-dependent cell death by causing dissociation of 14-3-3z from caspase-2 [506]. The SIRT1 activator SRT1720 increases metastasis of breast cancer cells to the lung irrespective of cisplatin treatment [451]. Renal injury induced by cisplatin could be ameliorated by SIRT1 activation, which reduced acetylation of the p65 subunit of NF-kB induced by cisplatin, thus lowering inflammation [507]. Many of the proteins involved in the effects of SIRT1 in cancer are depicted in Fig. 10.

SIRT2 in cancer Most evidence points to a role for SIRT2 as a tumor suppressor [435]. A protective role of SIRT2 against cancer is supported by its mitotic checkpoint function in the presence of mitotic stress, suggesting a role for SIRT2 activators in cancer therapy [41,146,508]. In gliomas, SIRT2 expression is reduced [509]. It may be that SIRT2 itself is lost, since its locus, at 19q13.2, is often deleted in gliomas [509]. SIRT2 inhibits colony formation in glioma cell lines [509]. This appears to involve an effect on microtubule regulation [509]. Overexpression of a SIRT2 phosphorylation site mutant in a glioblastoma cell line exposed to mitotic stress reduces hyperploid cells [510]. On the other hand, C6 glioma cells undergo apoptosis when SIRT2 is inhibited, pointing to a pathological role

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

of SIRT2 in this cancer, and possibly to a benefit from pharmacological inhibition of SIRT2 [511]. Similarly, HeLa cervical carcinoma cells undergo apoptosis in response to SIRT2 downregulation [462]. SIRT2 expression is also reduced in esophageal and gastric adenocarcinomas [512]. In melanoma, an inactivating mutation has been identified in the catalytic domain of SIRT2 [513]. The ability of SIRT2 to act as a tumor suppressor is supported by the development of hepatocellular carcinoma in male mice deficient in SIRT2 and mammary tumors in SIRT2–/– females [514]. SIRT2 is, moreover, reduced in human mammary and hepatocellular carcinomas [514]. SIRT2 regulates APC/cyclosome (a member of the RING family of ubiquitin ligases) by deacetylation of its coactivators APCCDH1 and CDC20 [514]. Loss of SIRT2 increases regulators of mitosis, including Aurora-A and Aurora-B that direct centrosome amplification, aneuploidy, and mitotic cell death. Further evidence that SIRT2 acts as a tumor suppressor is the hyperacetylation of the SIRT2 target lysine 53 of histone H3 in cancer cells [147]. In acute myeloid leukemia cells, however, SIRT2 and NAMPT are upregulated and participate in the aberrant proliferation and survival of leukemic cells [515]. SIRT3 in cancer The mitochondrial sirtuin SIRT3 regulates metabolism and oxidative stress [252]. There is some evidence that SIRT3 acts as a mitochondrial tumor suppressor [135]. Loss of SIRT3 results in an elevation in ROS that increases stabilization of HIF-a and thereby increases expression of HIF-dependent genes, metabolic reprogramming toward glycolysis, and a drive toward tumor phenotype [138]. But whether SIRT3 serves as a tumor suppressor or promoter is controversial [516]. For example, SIRT3 levels are reduced in breast cancer, consistent with it being a tumor suppressor [135], but in malignant, lymph node-positive, breast cancer SIRT3 is increased [517]. SIRT3 is downregulated in hepatocellular carcinoma tissue [518]. Adenovirus-mediated overexpression of SIRT3 inhibited hepatoceullar carcinoma cell growth and induced apoptosis [518]. This was associated with NAD þ suppression, reduction in ERK1/2 signaling, activation of AKT and JNK signaling, and, by downregulation of NDM2, results in reduced p53 degradation and thus increased p53 levels [518]. Oral squamous cell carcinoma cells exhibiting resistance to anoikis (a form of programmed cell death) have elevated SIRT3 expression, which may have arisen from reduced RIP [519]. Inhibition of SIRT3 inhibits anoikis resistance in this cancer and lowers tumor incidence [519]. The ability of SIRT3 to protect cells from oxidative stress is dependent on inhibition of the mitochondrial matrix protein IDH2, which is a major factor in cancer [520]. SIRT4 in cancer Photodamage induces upregulation of SIRT1 and SIRT4 in skin cells, followed by loss of sirtuin proteins, then later accumulation of SIRT4 in particular [445]. In squamous cell carcinoma, expression of all 7 sirtuin genes is increased [445]. SIRT6 in cancer SIRT6 appears to be a tumor suppressor. It deacetylates histones H3K9 and H3K56 [26–28] and is involved in DNA repair at double-stranded breaks [521,522]. H3K56 is hyperacetylated in skin, thyroid, breast, liver, and colon cancers [147]. Induction of SIRT6 overexpression in a variety of cancer cell lines results in massive apoptosis [523]. This involves its mono-ADP-

155

ribosyltransferase activity, but not its deacetylase activity, leading to activation of p53 and p73 apoptotic signaling cascades [523]. SIRT7 in cancer SIRT7 levels are elevated in breast cancer generally, including node-positive breast cancer [517] and thyroid cancer [435]. While SIRT7 might not be involved in cancer onset, it seems to be crucial for maintaining the cancer phenotype [524]. It does so by deacetylating acetylated histone H3K18 at lysine 18 [52]. Of 276 binding sites for SIRT7 in the genome, 74% are in the proximal promoter of various genes where H3K18 is found and, by SIRT7mediated deacetylation, the transcription of these is repressed [52]. This action of SIRT7 leads to anchorage-independent growth and escape from contact inhibition, consistent with progression of tumors to aggressive phenotypes and to poor prognosis. Moreover, depletion of SIRT7 markedly impedes tumorigenicity [52]. How might the dilemma be resolved? Normally SIRT1 responds to stress by promoting cell survival. This involves its ability to induce cell cycle arrest, DNA repair, and inhibition of apoptosis. But under extreme conditions SIRT1 [525], SIRT2 [143], and SIRT3 [526] act to protect the organism by inducing cell senescence or apoptosis. When overexpressed, SIRT1 inhibits senescence caused by oncogenes [527], and SIRT1 inhibition leads to senescence-like growth arrest [528]. So it seems that if the stress is chronic or the level of damage exceeds a certain threshold then SIRT1 may cause cell senescence. But when such chronic stress or DNA damage is accompanied by loss of a tumor repressor or loss of another checkpoint factor, then an imbalance might ensue, resulting in SIRT1 being overexpressed beyond a critical limit. By stimulating cell growth and inhibiting apoptosis this would contribute to transformation and tumorigenesis. Clearly, however, more research is needed to understand the role of sirtuins in cancer.

Inflammatory arthropathies While a role for sirtuins in inflammatory diseases has been known for a while, only more recently has it become apparent that sirtuins are involved in the acute inflammatory response as well. This involves a switch from increased glycolysis to increased fatty acid oxidation as early inflammation converts to late inflammation. SIRT6 reduces glycolysis, while SIRT1 increases fatty acid oxidation [529]. Mediators include PPAR-g coactivators PGC-1a and PGC-1b that increase CD36 in the external plasma membrane and, in mitochondria, the fatty acid transporter CPT1 [529]. Chronic inflammation is not only a major characteristic of autoimmune diseases, but is a general feature of most common diseases of aging. SIRT1 suppresses both innate and adaptive immunity. SIRT1 expression is highly induced in anergic T cells. The early response genes 2 and 3 bind to the SIRT1 promoter, synergize with FOXO3A, and activate SIRT1 transcription [530]. Tcell anergy can be reversed by IL-2, which, via activation of the PI3K–AKT pathways, sequesters FOXO3A, thus reducing SIRT1 transcription [530]. In regulatory T cells, SIRT1 deacetylates FOXP3, a protein crucial for ensuring a balanced immune response [531]. This leads to FOXP3 polyubiquination and proteosome-mediated degradation. TNF-a upregulates proinflammatory MMP-9, IL-1b, IL-6, and iNOS in NIH/3T3 fibroblasts [532]. Resveratrol, by activating SIRT1, ameliorates these effects by causing deacetylation of the NF-kB subunit RelA/p65 and attenuating phosphorylation of TOR and S6 ribosomal protein [532].

156

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

SIRT1 also deacetylates CIITA, a protein pivotal to MHC II activation and increased MHC II gene transcription in macrophages [533]. The activation and ensuing effects of SIRT1 can be triggered by NAMPT, which increases NAD þ [533]. SIRT1 is upregulated in synovial tissue and fibroblasts from patients with rheumatoid arthritis and appears to contribute to proinflammatory cytokine production, as well as in opposing apoptosis [534]. In osteoarthritis the proinflammatory mediator TNF-a induces cathepsin B-mediated cleavage of SIRT1 to inactivate it, and this action correlates with reduced cartilage-specific gene expression [535]. A key mediator of cartilage destruction in osteoarthritis is IL-1b. IL-1b induces NAMPT expression, which then activates SIRT1, leading to induction of ERK and p38 kinase activities that have a positive feedback effect on SIRT1 activity [536]. SIRT1–ERK is involved in IL-1b-induced chondrocyte dedifferentiation, which is mediated by SOX-9 [536]. Based on data from heterozygous SIRT1 knockout mice, SIRT1 may prolong the viability of articular chondrocytes [537]. The SIRT1 pathway is also involved in the cytoprotective and anti-inflammatory effects of melatonin, as shown in an oxidative stress-stimulated chondrocyte model of osteoarthritis [538]. Resveratrol has anti-inflammatory effects on articular chondrocytes stimulated with IL-1b and this involves SIRT1-mediated deacetylation of p65, followed by suppression of the nuclear translocation of NF-kB and a reduction in iNOS and NO [539]. The different roles of SIRT1 and roles for other sirtuins in inflammatory conditions clearly require more research.

Osteoporosis Activation of SIRT1 by resveratrol produces expression in mesenchymal stem cells of the bone-specific transcription factor RUNX2 [540,541]. Osteocalcin is also upregulated [540], whereas genes for adipo-lineage proteins PPAR-g [540,541] and leptin [540] are downregulated. This leads to spontaneous osteogenesis, mediated by an increase in SIRT1/FOXO3A complex formation and increased FOXO3A-dependent transcriptional activity, which includes binding of FOXO3A to a site in the RNX2 promoter [540]. PPAR-g expression is reduced as part of the activation and differentiation of mesenchymal stem cells to osteoblasts in osteogenic repair that occurs in response to bone injury [541]. SIRT1 contributes to maintenance of the stem cell pool by ensuring prematurity of hemopoietic stem cells via activation of FOXOs, inhibition of p53, and elimination of ROS [542]. SIRT1 regulates bone mass by inhibiting sclerostin [543]. Thus in various ways SIRT1 activation may help in the treatment of osteoporosis

Reproductive function SIRT1 affects fertility. A point mutation that disrupts SIRT1 activity, but with no effect on SIRT1 gene transcription, produces a milder phenotype than that seen in SIRT1 knockout mice [544]. Female mice were fertile rather than sterile, although male mice lacking either SIRT1 activity or transcription were sterile and hypermetabolic [544]. In human ovarian tissue the ability of resveratrol to increase mRNAs for SIRT1, lutenizing hormone receptor, steroidogenic acute regulatory protein, and P40 aromatase points to a role for SIRT1 in the luteal phase of the ovarian cycle [545]. Androgen depletion results in an increase in SIRT1 and eNOS expression in corpus cavernosum tissue [546]. The findings do not, however, appear to explain the effects of aging on structural changes in the corpus cavernosum [546].

Other medical conditions The critical role of CD8 T-cells in protection against viral infections involves SIRT1-mediated regulation of epigenetic remodeling and energy metabolism during promotion, by BATF, of CD8 T-cell differentiation [547]. In the lung, SIRT1 activation by SRT172 or elevated SIRT1 gene expression protects against emphysema via a FOXO3A-dependent mechanism [548]. In response to air pollution, SIRT1 prevents Kruppel-like factor 2-mediated expression of the thrombomodulin gene, thus lowering lung coagulation caused by exposure to air-born particulate matter [549]. Furthermore, a dimeric derivative of resveratrol, by upregulating SIRT1 and FOXO3A, can inhibit cigarette smoke-induced autophagy in human bronchial epithelial cells [550]. Inhibition of SIRT1 by ethanol is crucially involved in alcoholic fatty liver disease, and this involves overexpression of miR-217 [551]. miR-217 impairs SIRT1-regulated genes for lipogenic or fatty acid oxidation, as well as the important lipid regulator lipin1 [551]. By activating AMPK and SIRT1, resveratrol can prevent alcoholic fatty liver [552]. SIRT3 also ameliorates alcohol-induced mitochondrial hyperacetylation in a mouse model of alcoholic liver disease [553]. Skin damage by UV radiation and H2O2 involves downregulation of SIRT1 in human keratinocytes [554]. Resveratrol can reverse this process in a SIRT1-dependent manner. Resveratrol, by activating SIRT1 in skeletal muscle, helps prevent pathological changes caused by hind limb uploading in rats, a model of microgravity of space-flight [555]. Resveratrol is therefore a physical exercise mimetic. This finding adds to the observations noted earlier that resveratrol can increase oxidative type I muscle fibers and resistance to muscle fatigue, thus allowing mice to run twice as far before exhaustion [244]. A beneficial effect in tendinitis is also apparent, in that activation of SIRT1 inhibits inflammation and apoptosis in human tenocytes [556]. In the eye, SIRT1 preserves retinal ganglion cells during hypoxia [557]. The neuroprotective effect of SIRT1 involves an interaction with apoptotic signaling proteins to reduce hypoxiainduced apoptosis [557].

Aging Sirtuins protect against many of the various aging-associated conditions discussed above [19]. These conditions are often interrelated. For example, liver cancers can be induced by inflammation associated with the metabolic syndrome [19]. The role of SIRT1 during aging seems to involve the orchestration of different stress response pathways [558]. This involves targeting of multiple transcriptional regulators, such as p53, FOXO, and HSF1. SIRT1 activity is modulated by nutrient availability and declines with aging. The decline in SIRT1 in liver with aging is mediated by binding of repressive CCAT/enhancer binding protein/HDAC-1 complexes to the SIRT1 promoter [559]. The kinase TOR has an important role in aging. TOR is part of an evolutionarily conserved signaling pathway linking extracellular stimuli with intracellular processes such as cellular growth, metabolism, translational control, proliferation, and inhibition of autophagy [558]. Stress triggers protein misfolding and this depletes the pool of chaperones inside cells. TOR is able to sense and respond differentially to mild and severe depletion of different chaperones [560]. As a result, there is continuous integration of levels of nutrients available to the cell with protein homeostasis within the cell. The efficiency of the heat shock response pathway is impaired with aging. This leads to changes in

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

chaperone levels, and such changes adversely affect autophagosome clearance [558]. Thus the levels SIRT1 and TOR are central hubs of the stress response network connecting metabolism, autophagy, DNA damage, protein homeostasis, and heat shock response [558]. A characteristic feature of aging is telomere shortening. Telomeres protect the ends of chromosomes and shorten with each cell replication event [561]. Such shortening is opposed by telomerase. SIRT1 depletion leads to telomere dysfunction [562]. SIRT1 is a positive regulator of telomere length. In mice engineered to contain additional copies of the SIRT1 gene [243] telomere shortening with aging is attenuated [563]. SIRT1 interacts with the telomeric repeat sequences and promotes recombination at different chromosomal regions, including telomeres, centromeres, and chromosome arms. This effect is dependent on telomerase activity [563]. Aging may be a metabolic condition, since defective energy metabolism seems to be intimately intertwined with DNA damage. Dysfunction of telomere maintenance, via a decrease in PGC-1a and thence p53 activation, can affect mitochondrial biogenesis and function and thereby metabolic homeostasis [564]. As discussed earlier, SIRT1 serves as a NAD þ sensor that converts alterations in metabolic or redox state into adaptive transcriptional responses [17]. NAD þ is rate limiting in SIRT1mediated activation of PGC-1a, which coactivates transcription of nuclear-encoded mitochondrial genes [79]. The interplay between PARPs and sirtuins complements this mechanism [299]. PARP-1 binds DNA structures and nucleosomes, so affecting DNA repair, chromosome stability, and regulation of transcription [565]. PARP-1 uses NAD þ as a cosubstrate. NAD þ is converted to nicotinamide in generating polymers of ADP-ribose, which attach to acceptor proteins. When DNA is damaged strong stimulation of PARP-1 occurs, causing NAD þ levels to fall [565]. PARP-2 makes a smaller contribution. Thus deletion of PARP-1 or PARP-2 causes SIRT1 activation and thence mitochondrial biogenesis and fatty acid oxidation. During oxidative stress PARP-1 is activated, thus resulting in NAD þ consumption, meaning that less NAD þ is available for SIRT1 activity, so SIRT1 activity falls [75]. Thus by limiting NAD þ , PARP-1 is a gatekeeper for SIRT1 activity. The interaction between PARP-1 and SIRT1 affects not only metabolic regulation, but most likely aging. Mice deficient in PARP-1 or PARP-2 only exhibit DNA damage when exposed to genotoxic stress [566,567], and dual knockout of these PARPs is embryologically lethal, demonstrating that either alone can compensate for loss of the other [568]. It has long been known that maximal PARylation capacity arising from intrinsic PARP activity correlates with life span of multiple mammalian species [569]. With age, PARP activity of leukocytes decreases [569,570]. However, rather than high PARP activity being able to enhance life span, high PARP may be a consequence of longevity [299]. In support of this, high PARP-1 can in fact cause age-related diseases and premature death [300]. These mice exhibit elevated fat mass, body weight, and glucose intolerance. Thus perhaps low PARP activity might be prolongevity. The effect on life span of PARP deletion in mice differs, moreover, in different strains [75]. With age, endothelial-derived NO declines as a result of impaired eNOS activity and rising inactivation of NO caused by elevation in superoxide [571]. This leads to vascoconstriction, impaired perfusion, vascular inflammation, atherosclerosis, and impeded endothelial repair [572]. As discussed earlier, SIRT1 stimulates eNOS activity by deacetylating it, thus causing a rise in NO [109]. Moreover, during calorie restriction NO derived from eNOS increases and stimulates SIRT1 expression [355]. With aging, miR-34a [172,382] and miR-217 [573] increase and downregulate SIRT1 by posttranscriptional effects on SIRT1

157

mRNA stability, leading to endothelial senescence, apoptosis, and impaired angiogenic signaling. SIRT1 delays senescence of human bone marrow-derived and adipose-derived mesenchymal stem cells [574]. The effect of SIRT1 involves in part its ability to delay the proaging factor p16INK4A [574]. Glucose restriction of lung fibroblasts can serve as a calorie restriction mimetic [575]. This results in an increase in SIRT1 expression, activation of AKT/p70S6K1, histone deacetylation, and methylation of the INK4A promoter, decreasing p16INK4A expression, thus leading to chromatin remodeling and inhibition of cellular senescence [575]. Conversely, overexpression of p16INK4A leads to early replicative senescence [575]. Another calorie restriction mimetic, 2-deoxyglucose, has beneficial effects on human fibroblast life span by increasing NAD þ and SIRT1 activity [576]. As discussed earlier, SIRT1 is involved in maintenance of chromosome stability during mitosis. Thus the loss of SIRT1 activity that occurs during aging and tumorigenesis may lead to aneuploidy and genomic instability [38]. With age, the decline in SIRT1 activity and expression in skeletal muscle are accompanied by increased inflammation, oxidative stress, and reduction in ability to rebuild muscle after injury or in response to exercise [577]. This is yet another example of the role of SIRT1 in the decline in bodily function with aging. Overexpression of the sirtuin target PGC-1a in skeletal muscle increases life span by over 30% and counteracts molecular changes as well as conditions of aging [578]. SIRT1 expression is, moreover, increased, pointing to a positive feedback mechanism. An increase in SIRT1 expression is seen in mice with deletion of the Ras-guanine nucleotide exchange factor RasGRF1 [579]. These mice exhibit partial attenuation of aging and an increase in maximum life span by 20%, thus linking Ras signaling to life span [579]. A mouse model of aging in which senescence is accelerated exhibits neuronal deterioration accompanied by cognitive decline that is SIRT1 dependent and preventable by resveratrol and melatonin [580]. In this mouse model oxidative stress in the hippocampus could be ameliorated by testosterone, which stimulated SIRT1 expression and eNOS activity, leading to suppression of endothelial senescence [581]. Senescent endothelial cells promote neuronal senescence via a hormonal mechanism. The decline in hippocampal SIRT1 with aging could be ameliorated by calorie restriction of old rats, seemingly by a posttranscriptional mechanism [582]. In models of Alzheimers’ disease, activation of SIRT1 by resveratrol or overexpression of SIRT1 in the hippocampus reduces hippocampal degeneration and lowers cognitive deficit [408]. This is accompanied by a reduction in acetylation of PGC-1a and p53 [408]. It may be that in Alzheimer’s disease aerobic glycolysis in the mediotemporal lobe of the brain depletes NAD þ and thus, by lowering SIRT1 activity, contributes to amyloidogenesis [419]. Loss of heteromeric acetyl choline receptors in brain has been found to result in premature brain aging [583]. The associated cellular stress that results may upregulate SIRT1 expression as an adaptive response to counter neurodegeneration [583]. Aging is associated with a loss of wake neurons in the basal forebrain and brainstem coinciding with the progressive loss of SIRT1 and accumulation of lipofuscin [584]. SIRT1 appears to be a critical neuoprotectant for wake neurons early in life, but its loss with aging renders these neurons more vulnerable to metabolic insults. A reciprocal decrease in blood of SIRT1 and an increase in the SIRT1 mRNA regulator miR-34a represent accessible biomarkers of aging, and in particular of impending neuronal decline [585]. The DNA methylome differs between newborns and centenarians [586]. Global hypomethylation is seen in centenarian DNA,

158

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

and, in the case of regulatory DNA, the most hypomethylated sequences are in CpG island promoters. Of genes implicated in aging, differentially methylated regions occur in promoters of SIRT5 and SIRT7 [586]. Epigenetic factors could, moreover, have a role in life span. Nutrients such as folate, riboflavin, cobalamin, choline, betaine, and methionine that are components of onecarbon metabolism affect DNA methylation by regulating levels of S-adenosyl-L-methionine, a methyl group donor, and S-adenosylL-homocysteine, an inhibitor of DNA methylation enzymes [587]. It has been argued that SIRT1 mediates the effects of resveratrol on DNA methylation patterns [588,589]. The active metabolite of vitamin D recruits histone acetylases; resveratrol, by activating SIRT1, causes deacetylation, as well microRNA effects that are either oncogenic or tumor suppressing [587,590]. SIRT6, by mono-ADP ribosylating PARP-1, promotes DNA repair in response to oxidative stress [591]. This effect appears relevant to aging and age-related diseases. The regenerative potential of embryonic stem cells has potential application in rejuvenation research in aging. In mouse embryonic stem cells, SIRT1 appears to have an important role in priming the PTEN/JNK/FOXO1 pathway to respond to ROS [592]. Although mice expressing 3-fold higher levels of SIRT1 under the control of the mouse SIRT1 promoter do not live longer, they exhibit healthier ageing [19]. This includes improvements in glucose homeostasis, bone mineralization, fewer carcinomas and sarcomas, less DNA damage [19], and a reduction in molecular markers of aging such as p16INK4A [19], which is also causally involved in aging [593–595]. The various diseases of aging are reduced in frequency. SIRT1 activation also participates in the beneficial effects that calorie restriction confers. The AMPK/SIRT1–PGC-1a–PPAR axis that plays such a vital role in activating mitochondrial function declines in activity in older age, suggesting that a dominant negative regulatory mechanism is involved [596]. The decline in each component is accompanied by an increase in SMRT with age, and the ability of SMRT to negatively regulate mitochondrial function that promotes aging and aging-related disease suggests potential avenues for alleviating the decline in bodily function in general with aging [596].

Clinical molecular genetics Several SNPs in the SIRT1 gene have shown associations with a variety of factors and disease conditions. These include associations with SIRT1 gene expression [597], energy expenditure [244], body fat [598], body mass index [597,599,600], obesity [597], blood pressure [598], metabolic syndrome (which involves a coding variant) [124], a reduction in acute response in insulin to a glucose load [601], risk of type 2 diabetes [601], anxiety disorders [224], major depressive disorder [602], telomere length [603], and no difference [604] or an increase [603] in longevity. The minor allele of the same variant that conferred protection of telomere length was associated with longevity [603]. A lack of association with type 2 diabetes has also been observed, although in this Dutch Famine Birth Cohort the minor alleles of 2 of the 3 SNPs studied were associated with a lower diabetes prevalence in individuals who had been exposed to famine prenatally [605]. For Alzheimer’s disease, both association [606] or lack of association [607] have been reported. No association has been found with bipolar disorder [606]. In the case of the SIRT2 gene, a SNP was associated with increased risk of Alzheimer’s disease in cases that lacked the APOE e4 genotype [608].

In an Italian population a SIRT3 exon 3 SNP was associated with longevity [609], and, in exon 5, a VNTR polymorphism that downregulated SIRT3 expression was associated with reduced life span [610]. Subsequently more extensive studies have mostly failed to replicate associations at the SIRT3 locus, suggesting that any effect is at best weak [611]. An association of SIRT3 variants with carotid media-intima thickness has been reported [612]. An SNP in the SIRT6 gene was associated with carotid plaque, and interactions were found with SNPs in the SIRT1, SIRT3, and SIRT5 genes [613]. Of other genes in sirtuin pathways, the FOXO3A gene has proved to be of particular interest. Over 10 studies, beginning in 2008 [614], have shown that polymorphisms in the FOXO3A gene are associated with human longevity. Exonic polymorphisms do not explain the association [615], whereas 2 SNPs found to be associated with longevity are located in the 30 UTR, suggesting a mechanism involving differential effects of alleles of these on FOXOA mRNA stability [616]. A site in intron 2, where other genetic variants associated with longevity are located, contains enhancer sequences targeted by p53 [617]. p53 also binds to a site 4 kb upstream leading to transactivation of FOXO3A by p53, and also p73, in mouse liver [618]. The ability of p53 to transduce FOXO3A expression in response to DNA damage and environmental conditions such as nutrient deprivation, growth factor deprivation, and hypoxia may also involve promoter binding of other transcription factors, including E2F1 [619] and HIF1-a [620]. There is as well evidence for an indirect inhibitory effect of p53 during oxidative stress, via induction of serum- and glucocorticoid-inducible kinase 1 [621], or via a direct effect on the FOXO3A promoter [622]. p53 and FOXO3A interact at multiple levels, including physically [159,623], sharing common target genes [624,625], stabilization by FOXO3A of the p53 protein [626], and indirect p53 activation by FOXO3A via upregulation of p19ARF, which inhibits the p53 ubiquitin ligase MDM2 [627]. Clearly FOXO3A and p53 are part of a transcriptional regulatory network that involves sirtuins, with important roles in aging and diseases of aging.

Conclusion It can be seen from this extensive detailed review that sirtuins exert a wide repertoire of actions on intracellular processes. Apart from roles in adapting cellular physiology to changes in nutrient conditions, there is now compelling evidence for their role in common diseases of aging. The potential therefore exists for development of drugs that target one or other of the seven sirtuins, either directly or indirectly, for treatment of the seven deadly chronic conditions of aging that plague human society in the modern era. References [1] Frye, R. A. Phylogenetic classification of prokaryotic and eukaryotic Sir2-like proteins. Biochem. Biophys. Res. Commun. 273:793–798; 2000. [2] Webster, B. R.; Lu, Z.; Sack, M. N.; Scott, I. The role of sirtuins in modulating redox stressors. Free Radic. Biol. Med. 52:281–290; 2012. [3] Baur, J. A.; Ungvari, Z.; Minor, R. K.; Le Couteur, D. G.; de Cabo, R. Are sirtuins viable targets for improving healthspan and lifespan? Nat. Rev. Drug Discov. 11:443–461; 2012. [4] Osborne, T. B.; Mendel, L. B.; Ferry, E. L. The effect of retardation of growth upon the breeding period and duration of life of rats. Science 45:294–295; 1917. [5] McCay, C. M.; Crowell, M. F.; Maynard, L. A. The effect of retarded growth upon the length of life span and upon the ultimate body size. 1935. Nutrition 5:155–171; 1935. [6] Howitz, K. T.; Bitterman, K. J.; Cohen, H. Y.; Lamming, D. W.; Lavu, S.; Wood, J. G.; Zipkin, R. E.; Chung, P.; Kisielewski, A.; Zhang, L. L.; Scherer, B.; Sinclair, D. A. Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature 425:191–196; 2003.

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

[7] Bauer, J. H.; Goupil, S.; Garber, G. B.; Helfand, S. L. An accelerated assay for the identification of lifespan-extending interventions in Drosophila melanogaster. Proc. Natl. Acad. Sci. USA 101:12980–12985; 2004. [8] Wood, J. G.; Rogina, B.; Lavu, S.; Howitz, K.; Helfand, S. L.; Tatar, M.; Sinclair, D. Sirtuin activators mimic caloric restriction and delay ageing in metazoans. Nature 430:686–689; 2004. [9] Olshansky, S. J.; Carnes, B. A.; Cassel, C. In search of Methuselah: estimating the upper limits to human longevity. Science 250:634–640; 1990. [10] Ingram, D. K.; Zhu, M.; Mamczarz, J.; Zou, S.; Lane, M. A.; Roth, G. S.; deCabo, R. Calorie restriction mimetics: an emerging research field. Aging Cell 5:97–108; 2006. [11] Fries, J. F. Aging, natural death, and the compression of morbidity. N. Engl. J. Med. 303:130–135; 1980. [12] Shore, D.; Squire, M.; Nasmyth, K. A. Characterization of two genes required for the position-effect control of yeast mating-type genes. EMBO J 3:2817–2823; 1984. [13] Sinclair, D. A.; Guarente, L. Extrachromosomal rDNA circles—a cause of aging in yeast. Cell 91:1033–1042; 1997. [14] Tissenbaum, H. A.; Guarente, L. Increased dosage of a sir-2 gene extends lifespan in Caenorhabditis elegans. Nature 410:227–230; 2001. [15] Rogina, B.; Helfand, S. L. Sir2 mediates longevity in the fly through a pathway related to calorie restriction. Proc. Natl. Acad. Sci. USA 101:15998–16003; 2004. [16] Imai, S.; Armstrong, C. M.; Kaeberlein, M.; Guarente, L. Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature 403:795–800; 2000. [17] Guarente, L. Sir2 links chromatin silencing, metabolism, and aging. Genes Dev. 14:1021–1026; 2000. [18] Herranz, D.; Serrano, M. SIRT1: recent lessons from mouse models. Nat. Rev. Cancer 10:819–823; 2010. ˜oz-Martin, M.; Can ˜amero, M.; Mulero, F.; Martinez-Pastor, B.; [19] Herranz, D.; Mun Fernandez-Capetillo, O.; Serrano, M. Sirt1 improves healthy ageing and protects from metabolic syndrome-associated cancer. Nat. Commun 1:3; 2010. [20] Viswanathan, M.; Guarente, L. Regulation of Caenorhabditis elegans lifespan by sir-2.1 transgenes. Nature 477:E1–E2; 2011. [21] Rizki, G.; Iwata, T. N.; Li, J.; Riedel, C. G.; Picard, C. L.; Jan, M.; Murphy, C. T.; Lee, S. S. The evolutionarily conserved longevity determinants HCF-1 and SIR-2.1/SIRT1 collaborate to regulate DAF-16/FOXO. PLoS Genet. 7:e1002235; 2011. [22] Burnett, C.; Valentini, S.; Cabreiro, F.; Goss, M.; Somogyva´ri, M.; Piper, M. D.; Hoddinott, M.; Sutphin, G. L.; Leko, V.; McElwee, J. J.; Vazquez-Manrique, R. P.; Orfila, A. M.; Ackerman, D.; Au, C.; Vinti, G.; Riesen, M.; Howard, K.; Neri, C.; Bedalov, A.; Kaeberlein, M.; Soti, C.; Partridge, L.; Gems, D. Absence of effects of Sir2 overexpression on lifespan in C. elegans and Drosophila. Nature 477:482–485; 2011. [23] Mattison, J. A.; Roth, G. S.; Beasley, M. T.; Tilmont, E. M.; Handy, A. M.; Herbert, R. L.; Longo, D. L.; Allison, D. B.; Young, J. E.; Bryant, M.; Barnard, D.; Ward, W. F.; Qi, W.; Ingram, D. K.; de Cabo, R. Impact of caloric restriction on health and survival in rhesus monkeys from the NIA study. Nature 489:318–321; 2012. [24] Colman, R. J.; Anderson, R. M.; Johnson, S. C.; Kastman, E. K.; Kosmatka, K. J.; Beasley, T. M.; Allison, D. B.; Cruzen, C.; Simmons, H. A.; Kemnitz, J. W.; Weindruch, R. Caloric restriction delays disease onset and mortality in rhesus monkeys. Science 325:201–204; 2009. [25] Kanfi, Y.; Naiman, S.; Amir, G.; Peshti, V.; Zinman, G.; Nahum, L.; Bar-Joseph, Z.; Cohen, H. Y. The sirtuin SIRT6 regulates lifespan in male mice. Nature 483:218–221; 2012. [26] Michishita, E.; McCord, R. A.; Berber, E.; Kioi, M.; Padilla-Nash, H.; Damian, M.; Cheung, P.; Kusumoto, R.; Kawahara, T. L.; Barrett, J. C.; Chang, H. Y.; Bohr, V. A.; Ried, T.; Gozani, O.; Chua, K. F. SIRT6 is a histone H3 lysine 9 deacetylase that modulates telomeric chromatin. Nature 452:492–496; 2008. [27] Yang, B.; Zwaans, B. M.; Eckersdorff, M.; Lombard, D. B. The sirtuin SIRT6 deacetylates H3 K56Ac in vivo to promote genomic stability. Cell Cycle 8:2662–2663; 2009. [28] Michishita, E.; McCord, R. A.; Boxer, L. D.; Barber, M. F.; Hong, T.; Gozani, O.; Chua, K. F. Cell cycle-dependent deacetylation of telomeric histone H3 lysine K56 by human SIRT6. Cell Cycle 8:2664–2666; 2009. [29] Liao, C. Y.; Kennedy, B. K. Will the real aging Sirtuin please stand up? Cell Res. 22:1215–1217; 2012. [30] Swindell, W. R. Genes and gene expression modules associated with caloric restriction and aging in the laboratory mouse. BMC Genomics 10:585; 2009. [31] Estep 3rd P. W.; Warner, J. B.; Bulyk, M. L. Short-term calorie restriction in male mice feminizes gene expression and alters key regulators of conserved aging regulatory pathways. PLoS One 4:e5242; 2009. [32] Mostoslavsky, R.; Chua, K. F.; Lombard, D. B.; Pang, W. W.; Fischer, M. R.; Gellon, L.; Liu, P.; Mostoslavsky, G.; Franco, S.; Murphy, M. M.; Mills, K. D.; Patel, P.; Hsu, J. T.; Hong, A. L.; Ford, E.; Cheng, H. L.; Kennedy, C.; Nunez, N.; Bronson, R.; Frendewey, D.; Auerbach, W.; Valenzuela, D.; Karow, M.; Hottiger, M. O.; Hursting, S.; Barrett, J. C.; Guarente, L.; Mulligan, R.; Demple, B.; Yancopoulos, G. D.; Alt, F. W. Genomic instability and aginglike phenotype in the absence of mammalian SIRT6. Cell 124:315–329; 2006. [33] Canto, C.; Auwerx, J. Don’t write sirtuins off. Nature 477:411; 2011. [34] Guarente, L.; Franklin, H. Epstein lecture: sirtuins, aging, and medicine. N. Engl. J. Med. 364:2235–2244; 2011.

159

[35] Nogueiras, R.; Habegger, K. M.; Chaudhary, N.; Finan, B.; Banks, A. S.; ¨ M. H. Dietrich, M. O.; Horvath, T. L.; Sinclair, D. A.; Pfluger, P. T.; Tschop, Sirtuin 1 and sirtuin 3: physiological modulators of metabolism. Physiol. Rev. 92:1479–1514; 2012. [36] Michishita, E.; Park, J. Y.; Burneskis, J. M.; Barrett, J. C.; Horikawa, I. Evolutionarily conserved and nonconserved cellular localizations and functions of human SIRT proteins. Mol. Biol. Cell 16:4623–4635; 2005. [37] Tanno, M.; Sakamoto, J.; Miura, T.; Shimamoto, K.; Horio, Y. Nucleocytoplasmic shuttling of the NAD þ -dependent histone deacetylase SIRT1. J. Biol. Chem. 282:6823–6832; 2007. [38] Fatoba, S. T.; Okorokov, A. L. Human SIRT1 associates with mitotic chromatin and contributes to chromosomal condensation. Cell Cycle. 10:2317–2322; 2011. [39] North, B. J.; Marshall, B. L.; Borra, M. T.; Denu, J. M.; Verdin, E. The human Sir2 ortholog, SIRT2, is an NAD þ -dependent tubulin deacetylase. Mol. Cell 11:437–444; 2003. [40] Jing, E.; Gesta, S.; Kahn, C. R. SIRT2 regulates adipocyte differentiation through FoxO1 acetylation/deacetylation. Cell Metab. 6:105–114; 2007. [41] Dryden, S. C.; Nahhas, F. A.; Nowak, J. E.; Goustin, A. S.; Tainsky, M. A. Role for human SIRT2 NAD-dependent deacetylase activity in control of mitotic exit in the cell cycle. Mol. Cell. Biol. 23:3173–3185; 2003. [42] Lu, L.; Li, L.; Lv, X.; Wu, X. S.; Liu, D. P.; Liang, C. C. Modulations of hMOF autoacetylation by SIRT1 regulate hMOF recruitment and activities on the chromatin. Cell Res. 21:1182–1195; 2011. [43] Vaquer, A.; Scher, M. B.; Lee, D. H.; Sutton, A.; Cheng, H. L.; Alt, F. W.; Serrano, L.; Sternglanz, R.; Reinberg, D. SirT2 is a histone deacetylase with preference for histone H4 Lys 16 during mitosis. Genes Dev. 20:1256–1261; 2006. [44] Lampson, M. A.; Kapoor, T. M. The human mitotic checkpoint protein BubR1 regulates chromosome-spindle attachments. Nat. Cell Biol. 7:93–98; 2005. [45] He, W.; Newman, J. C.; Wang, M. Z.; Ho, L.; Verdin, E. Mitochondrial sirtuins: regulators of protein acylation and metabolism. Trends Endocrinol. Metab. ; August 16, 2012. [Epub ahead of print]. [46] Ardestani, P. M.; Liang, F. Sub-cellular localization, expression and functions of Sirt6 during the cell cycle in HeLa cells. Nucleus 3:442–451; 2012. [47] Milne, J. C.; Denu, J. M. The sirtuin family: therapeutic targets to treat diseases of aging. Curr. Opin. Chem. Biol. 12:11–17; 2008. [48] Donmez, G.; Guarente, L. Aging and disease: connections to sirtuins. Aging Cell 9:285–290; 2010. [49] Haigis, M. C.; Sinclair, D. A. Mammalian sirtuins: biological insights and disease relevance. Annu. Rev. Pathol 5:253–295; 2010. [50] Vaziri, H.; Dessain, S. K.; Ng-Eaton, E.; Imai, S. I.; Frye, R. A.; Pandita, T. K.; Guarente, L.; Weinberg, R. A. hSIR2SIRT1 functions as an NAD-dependent p53 deacetylase. Cell 107:149–159; 2001. [51] Vakhrusheva, O.; Smolka, C.; Gajawada, P.; Kostin, S.; Boettger, T.; Kubin, T.; Braun, T.; Bober, E. Sirt7 increases stress resistance of cardiomyocytes and prevents apoptosis and inflammatory cardiomyopathy in mice. Circ. Res. 102:703–710; 2008. [52] Barber, M. F.; Michishita-Kioi, E.; Xi, Y.; Tasselli, L.; Kioi, M.; Moqtaderi, Z.; Tennen, R. I.; Paredes, S.; Young, N. L.; Chen, K.; Struhl, K.; Garcia, B. A.; Gozani, O.; Li, W.; Chua, K. F. SIRT7 links H3K18 deacetylation to maintenance of oncogenic transformation. Nature 487:114–118; 2012. [53] Haigis, M. C.; Mostoslavsky, R.; Haigis, K. M.; Fahie, K.; Christodoulou, D. C.; Murphy, A. J.; Valenzuela, D. M.; Yancopoulos, G. D.; Karow, M.; Blander, G.; Wolberger, C.; Prolla, T. A.; Weindruch, R.; Alt, F. W.; Guarente, L. SIRT4 inhibits glutamate dehydrogenase and opposes the effects of calorie restriction in pancreatic beta cells. Cell 126:941–954; 2006. [54] Liszt, G.; Ford, E.; Kurtev, M.; Guarente, L. Mouse Sir2 homolog SIRT6 is a nuclear ADP-ribosyltransferase. J. Biol. Chem. 280:21313–21320; 2005. [55] Shi, T.; Wang, F.; Stieren, E.; Tong, Q. SIRT3, a mitochondrial sirtuin deacetylase, regulates mitochondrial function and thermogenesis in brown adipocytes. J. Biol. Chem. 280:13560–13567; 2005. [56] Zhong, L.; D’Urso, A.; Toiber, D.; Sebastian, C.; Henry, R. E.; Vadysirisack, D. D.; Guimaraes, A.; Marinelli, B.; Wikstrom, J. D.; Nir, T.; Clish, C. B.; Vaitheesvaran, B.; Iliopoulos, O.; Kurland, I.; Dor, Y.; Weissleder, R.; Shirihai, O. S.; Ellisen, L. W.; Espinosa, J. M.; Mostoslavsky, R. The histone deacetylase Sirt6 regulates glucose homeostasis via Hif1a. Cell 140:280–293; 2010. [57] Nakagawa, T.; Lomb, D. J.; Haigis, M. C.; Guarente, L. SIRT5 deacetylates carbamoyl phosphate synthetase 1 and regulates the urea cycle. Cell 137:560–570; 2009. [58] Du, J.; Zhou, Y.; Su, X.; Yu, J. J.; Khan, S.; Jiang, H.; Kim, J.; Woo, J.; Kim, J. H.; Choi, B. H.; He, B.; Chen, W.; Zhang, S.; Cerione, R. A.; Auwerx, J.; Hao, Q.; Lin, H. Sirt5 is a NAD-dependent protein lysine demalonylase and desuccinylase. Science 334:806–809; 2011. [59] Peng, C.; Lu, Z.; Xie, Z.; Cheng, Z.; Chen, Y.; Tan, M.; Luo, H.; Zhang, Y.; He, W.; Yang, K.; Zwaans, B. M.; Tishkoff, D.; Ho, L.; Lombard, D.; He, T. C.; Dai, J.; Verdin, E.; Ye, Y.; Zhao, Y. The first identification of lysine malonylation substrates and its regulatory enzyme. Mol. Cell. Proteomics. 10:M111.012658; 2011. [60] Kang, H.; Suh, J. Y.; Jung, Y. S.; Jung, J. W.; Kim, M. K.; Chung, J. H. Peptide switch is essential for Sirt1 deacetylase activity. Mol. Cell 44:203–213; 2011. [61] Pan, M.; Yuan, H.; Brent, M.; Ding, E. C.; Marmorstein, R. SIRT1 contains Nand C-terminal regions that potentiate deacetylase activity. J. Biol. Chem. 287:2468–2476; 2012.

160

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

[62] Chen, D.; Bruno, J.; Easlon, E.; Lin, S. J.; Cheng, H. L.; Alt, F. W.; Guarente, L. Tissue-specific regulation of SIRT1 by calorie restriction. Genes Dev. 22:1753–1757; 2008. [63] Canto´, C.; Auwerx, J. Targeting sirtuin 1 to improve metabolism: all you need is NAD þ ? Pharmacol. Rev. 64:166–187; 2012. [64] Canto´, C.; Gerhart-Hines, Z.; Feige, J. N.; Lagouge, M.; Noriega, L.; Milne, J. C.; Elliott, P. J.; Puigserver, P.; Auwerx, J. AMPK regulates energy expenditure by modulating NAD þ metabolism and SIRT1 activity. Nature 458:1056–1060; 2009. [65] Kim, H. J.; Kim, J. H.; Noh, S.; Hur, H. J.; Sung, M. J.; Hwang, J. T.; Park, J. H.; Yang, H. J.; Kim, M. S.; Kwon, D. Y.; Yoon, S. H. Metabolomic analysis of livers and serum from high-fat diet induced obese mice. J. Proteome Res. 10:722–731; 2011. [66] Houtkooper, R. H.; Canto´, C.; Wanders, R. J.; Auwerx, J. The secret life of NAD þ : an old metabolite controlling new metabolic signaling pathways. Endocr. Rev. 31:194–223; 2010. [67] Collins, P. B.; Chaykin, S. The management of nicotinamide and nicotinic acid in the mouse. J. Biol. Chem. 247:778–783; 1972. [68] Bieganowski, P.; Brenner, C. Discoveries of nicotinamide riboside as a nutrient and conserved NRK genes establish a Preiss-Handler independent route to NAD þ in fungi and humans. Cell 117:495–502; 2004. [69] Yoshino, J.; Mills, K. F.; Yoon, M. J.; Imai, S. Nicotinamide mononucleotide, a key NAD þ intermediate, treats the pathophysiology of diet- and ageinduced diabetes in mice. Cell Metab. 14:528–536; 2011. [70] Bitterman, K. J.; Anderson, R. M.; Cohen, H. Y.; Latorre-Esteves, M.; Sinclair, D. A. Inhibition of silencing and accelerated aging by nicotinamide, a putative negative regulator of yeast sir2 and human SIRT1. J. Biol. Chem. 277:45099–45107; 2002. [71] Anderson, R. M.; Bitterman, K. J.; Wood, J. G.; Medvedik, O.; Sinclair, D. A. Nicotinamide and PNC1 govern lifespan extension by calorie restriction in Saccharomyces cerevisiae. Nature 423:181–189; 2003. [72] Yang, T.; Sauve, A. A. NAD metabolism and sirtuins: metabolic regulation of protein deacetylation in stress and toxicity. AAPS J 8:E632–E643; 2006. [73] Jang, S. Y.; Kang, H. T.; Hwang, E. S. Nicotinamide-induced mitophagy: event mediated by high NAD þ /NADH ratio and SIRT1 protein activation. J. Biol. Chem. 287:19304–19314; 2012. ¨ [74] Gerhart-Hines, Z.; Dominy Jr J. E.; Blattler, S. M.; Jedrychowski, M. P.; Banks, A. S.; Lim, J. H.; Chim, H.; Gyg, i. S. P.; Puigserver, P. The cAMP/PKA pathway rapidly activates SIRT1 to promote fatty acid oxidation independently of changes in NAD þ . Mol. Cell 44:851–863; 2011. [75] Bai, P.; Canto, C.; Oudart, H.; Brunya´nszki, A.; Cen, Y.; Thomas, C.; Yamamoto, H.; Huber, A.; Kiss, B.; Houtkooper, R. H.; Schoonjans, K.; Schreiber, V.; Sauve, A. A.; Menissier-de Murcia, J.; Auwerx, J. PARP-1 inhibition increases mitochondrial metabolism through SIRT1 activation. Cell Metab. 13:461–468; 2011. [76] Bai, P.; Canto´, C.; Brunya´nszki, A.; Huber, A.; Sza´nto´, M.; Cen, Y.; Yamamoto, H.; Houten, S. M.; Kiss, B.; Oudart, H.; Gergely, P.; Menissier-de Murcia, J.; Schreiber, V.; Sauve, A. A.; Auwerx, J. PARP-2 regulates SIRT1 expression and whole-body energy expenditure. Cell Metab. 13:450–460; 2011. [77] Barbosa, M. T.; Soares, S. M.; Novak, C. M.; Sinclair, D.; Levine, J. A.; Aksoy, P.; Chini, E. N. The enzyme CD38 (a NAD glycohydrolase, EC 3.2.2.5) is necessary for the development of diet-induced obesity. FASEB J 21:3629–3639; 2007. [78] Braunstein, M.; Sobel, R. E.; Allis, C. D.; Turner, B. M.; Broach, J. R. Efficient transcriptional silencing in Saccharomyces cerevisiae requires a heterochromatin histone acetylation pattern. Mol. Cell. Biol. 16:4349–4356; 1996. [79] Rodgers, J. T.; Lerin, C.; Haas, W.; Gygi, S. P.; Spiegelman, B. M.; Puigserver, P. Nutrient control of glucose homeostasis through a complex of PGC-1a and SIRT1. Nature 434:113–118; 2005. [80] Brunet, A.; Sweeney, L. B.; Sturgill, J. F.; Chua, K. F.; Greer, P. L.; Lin, Y.; Tran, H.; Ross, S. E.; Mostoslavsky, R.; Cohen, H. Y.; Hu, L. S.; Cheng, H. L.; Jedrychowski, M. P.; Gygi, S. P.; Sinclair, D. A.; Alt, F. W.; Greenberg, M. E. Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science 303:2011–2015; 2004. [81] van der Horst, A.; Tertoolen, L. G.; de Vries-Smits, L. M.; Frye, R. A.; Medema, R. H.; Burgering, B. M. FOXO4 is acetylated upon peroxide stress and deacetylated by the longevity protein hSir2SIRT1. J. Biol. Chem. 279:28873–28879; 2004. [82] Motta, M. C.; Divecha, N.; Lemieux, M.; Kamel, C.; Chen, D.; Gu, W.; Bultsma, Y.; McBurney, M.; Guarente, L. Mammalian SIRT1 represses forkhead transcription factors. Cell 116:551–563; 2004. [83] Purushotham, A.; Schug, T. T.; Xu, Q.; Surapureddi, S.; Guo, X.; Li, X. Hepatocyte-specific deletion of SIRT1 alters fatty acid metabolism and results in hepatic steatosis and inflammation. Cell Metab. 9:327–338; 2009. [84] Grimm, A. A.; Brace, C. S.; Wang, T.; Stormo, G. D.; Imai, S. A nutrientsensitive interaction between Sirt1 and HNF-1a regulates Crp expression. Aging Cell 10:305–317; 2011. [85] Gross, D. N.; van den Heuvel, A. P.; Birnbaum, M. J. The role of FoxO in the regulation of metabolism. Oncogene 27:2320–2336; 2008. [86] Wu, Z.; Puigserver, P.; Andersson, U.; Zhang, C.; Adelmant, G.; Mootha, V.; Troy, A.; Cinti, S.; Lowell, B.; Scarpulla, R. C.; Spiegelman, B. M. Mechanisms controlling mitochondrial biogenesis and respiration through the thermogenic coactivator PGC-1. Cell 98:115–124; 1999. [87] Hardie, D. G. AMP-activated/SNF1 protein kinases: conserved guardians of cellular energy. Nat. Rev. Mol. Cell. Biol. 8:774–785; 2007.

¨ [88] Jager, S.; Handschin, C.; St-Pierre, J.; Spiegelman, B. M. AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1a. Proc. Natl. Acad. Sci. USA 104:12017–12022; 2007. [89] Nemoto, S.; Fergusson, M. M.; Finkel, T. SIRT1 functionally interacts with the metabolic regulator and transcriptional coactivator PGC-1a. J. Biol. Chem. 280:16456–16460; 2005. [90] Greer, E. L.; Oskoui, P. R.; Banko, M. R.; Maniar, J. M.; Gygi, M. P.; Gygi, S. P.; Brunet, A. The energy sensor AMP-activated protein kinase directly regulates the mammalian FOXO3 transcription factor. J. Biol. Chem. 282:30107–30119; 2007. [91] Chau, M. D.; Gao, J.; Yang, Q.; Wu, Z.; Gromada, J. Fibroblast growth factor 21 regulates energy metabolism by activating the AMPK-SIRT1-PGC-1a pathway. Proc. Natl. Acad. Sci. USA 107:12553–12558; 2010. [92] Iwabu, M.; Yamauchi, T.; Okada-Iwabu, M.; Sato, K.; Nakagawa, T.; Funata, M.; Yamaguchi, M.; Namiki, S.; Nakayama, R.; Tabata, M.; Ogata, H.; Kubota, N.; Takamoto, I.; Hayashi, Y. K.; Yamauchi, N.; Waki, H.; Fukayama, M.; Nishino, I.; Tokuyama, K.; Ueki, K.; Oike, Y.; Ishii, S.; Hirose, K.; Shimizu, T.; Touhara, K.; Kadowaki, T. Adiponectin and AdipoR1 regulate PGC-1a and mitochondria by Ca2 þ and AMPK/SIRT1. Nature 464:1313–1319; 2010. [93] Li, L.; Pan, R.; Li, R.; Niemann, B.; Aurich, A. C.; Chen, Y.; Rohrbach, S. Mitochondrial biogenesis and peroxisome proliferator-activated receptor-g coactivator-1a (PGC-1a) deacetylation by physical activity: intact adipocytokine signaling is required. Diabetes 60:157–167; 2011. [94] Liu, Y.; Dentin, R.; Chen, D.; Hedrick, S.; Ravnskjaer, K.; Schenk, S.; Milne, J.; Meyers, D. J.; Cole, P.; Yates, J., 3rd; Olefsky, J.; Guarente, L.; Montminy, M. A fasting inducible switch modulates gluconeogenesis via activator/coactivator exchange. Nature 456:269–273; 2008. [95] Altarejos, J. Y.; Montminy, M. CREB and the CRTC co-activators: sensors for hormonal and metabolic signals. Nat. Rev. Mol. Cell. Biol. 12:141–151; 2011. [96] Mair, W.; Morantte, I.; Rodrigues, A. P.; Manning, G.; Montminy, M.; Shaw, R. J.; Dillin, A. Lifespan extension induced by AMPK and calcineurin is mediated by CRTC-1 and CREB. Nature 470:404–408; 2011. [97] Picard, F.; Kurtev, M.; Chung, N.; Topark-Ngarm, A.; Senawong, T.; Machado De Oliveira, R.; Leid, M.; McBurney, M. W.; Guarente, L. Sirt1 promotes fat mobilization in white adipocytes by repressing PPAR-g. Nature 429:771–776; 2004. [98] Heikkinen, S.; Auwerx, J.; Argmann, C. A. PPARg in human and mouse physiology. Biochim. Biophys. Acta 1771:999–1013; 2007. [99] Ponugoti, B.; Kim, D. H.; Xiao, Z.; Smith, Z.; Miao, J.; Zang, M.; Wu, S. Y.; Chiang, C. M.; Veenstra, T. D.; Kemper, J. K. SIRT1 deacetylates and inhibits SREBP-1C activity in regulation of hepatic lipid metabolism. J. Biol. Chem. 285:33959–33970; 2010. [100] Walker, A. K.; Yang, F.; Jiang, K.; Ji, J. Y.; Watts, J. L.; Purushotham, A.; Boss, O.; Hirsch, M. L.; Ribich, S.; Smith, J. J.; Israelian, K.; Westphal, C. H.; Rodgers, J. T.; Shioda, T.; Elson, S. L.; Mulligan, P.; Najafi-Shoushtari, H.; Black, J. C.; Thakur, J. K.; Kadyk, L. C.; Whetstine, J. R.; Mostoslavsky, R.; ¨ A. M. Conserved role of Puigserver, P.; Li, X.; Dyson, N. J.; Hart, A. C.; Na¨ ar, SIRT1 orthologs in fasting-dependent inhibition of the lipid/cholesterol regulator SREBP. Genes Dev. 24:1403–1417; 2010. [101] Asher, G.; Schibler, U. Crosstalk between components of circadian and metabolic cycles in mammals. Cell Metab 13:125–137; 2011. [102] Nakagawa, T.; Guarente, L. Sirtuins at a glance. J. Cell Sci. 124(Pt 6):833–838; 2011. [103] Ghosh, H. S.; Reizis, B.; Robbins, P. D. SIRT1 associates with eIF2-alpha and regulates the cellular stress response. Sci. Rep. 1(article 150):1–9; 2012. [104] Vaquero, A.; Scher, M.; Lee, D.; Erdjument-Bromage, H.; Tempst, P.; Reinberg, D. Human SirT1 interacts with histone H1 and promotes formation of facultative heterochromatin. Mol. Cell 16:93–105; 2004. [105] Vaquero, A.; Scher, M.; Erdjument-Bromage, H.; Tempst, P.; Serrano, L.; Reinberg, D. SIRT1 regulates the histone methyl-transferase SUV39H1 during heterochromatin formation. Nature 450:440–444; 2007. [106] He, H.; Yu, F. X.; Sun, C.; Luo, Y. CBP/p300 and SIRT1 are involved in transcriptional regulation of S-phase specific histone genes. PLoS One 6:e22088; 2011. [107] Bosch-Presegue´, L.; Raurell-Vila, H.; Marazuela-Duque, A.; Kane-Goldsmith, N.; Valle, A.; Oliver, J.; Serrano, L.; Vaquero, A. Stabilization of Suv39H1 by SirT1 is part of oxidative stress response and ensures genome protection. Mol. Cell 42:210–223; 2010. [108] Hallows, W. C.; Lee, S.; Denu, J. M. Sirtuins deacetylate and activate mammalian acetyl-CoA synthetases. Proc. Natl. Acad. Sci. USA 103:10230–10235; 2006. [109] Mattagajasingh, I.; Kim, C. S.; Naqvi, A.; Yamamori, T.; Hoffman, T. A.; Jung, S. B.; DeRicco, J.; Kasuno, K.; Irani, K. SIRT1 promotes endotheliumdependent vascular relaxation by activating endothelial nitric oxide synthase. Proc. Natl. Acad. Sci. USA 104:14855–14860; 2007. [110] Lee, I. H.; Cao, L.; Mostoslavsky, R.; Lombard, D. B.; Liu, J.; Bruns, N. E.; Tsokos, M.; Alt, F. W.; Finkel, T. A role for the NAD-dependent deacetylase Sirt1 in the regulation of autophagy. Proc. Natl. Acad. Sci. USA 105: 3374–3379; 2008. [111] Bernier, M.; Paul, R. K.; Martin-Montalvo, A.; Scheibye-Knudsen, M.; Song, S.; He, H. J.; Armour, S. M.; Hubbard, B. P.; Bohr, V. A.; Wang, L.; Zong, Y.; Sinclair, D. A.; de Cabo, R. Negative regulation of STAT3 protein-mediated cellular respiration by SIRT1 protein. J. Biol. Chem. 286:19270–19279; 2011. [112] Ferber, E. C.; Peck, B.; Delpuech, O.; Bell, G. P.; East, P.; Schulze, A. FOXO3a regulates reactive oxygen metabolism by inhibiting mitochondrial gene expression. Cell Death Differ. 19:968–979; 2012.

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

[113] Yeung, F.; Hoberg, J. E.; Ramsey, C. S.; Keller, M. D.; Jones, D. R.; Frye, R. A.; Mayo, M. W. Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J 23:2369–2380; 2004. [114] Mao, Z.; Hine, C.; Tian, X.; Van Meter, M.; Au, M.; Vaidya, A.; Seluanov, A.; Gorbunova, V. SIRT6 promotes DNA repair under stress by activating PARP1. Science 332:1443–1446; 2011. [115] Mao, Z.; Tian, X.; Van Meter, M.; Ke, Z.; Gorbunova, V.; Seluanov, A. Sirtuin 6 (SIRT6) rescues the decline of homologous recombination repair during replicative senescence. Proc. Natl. Acad. Sci. USA 109:11800–11805; 2012. [116] Schwer, B.; Schumacher, B.; Lombard, D. B.; Xiao, C.; Kurtev, M. V.; Gao, J.; Schneider, J. I.; Chai, H.; Bronson, R. T.; Tsai, L. H.; Deng, C. X.; Alt, F. W. Neural sirtuin 6 (Sirt6) ablation attenuates somatic growth and causes obesity. Proc. Natl. Acad. Sci. USA 107:21790–21794; 2010. [117] Kawahara, T. L.; Michishit, E.; Adler, A. S.; Damian, M.; Berbe, E.; Lin, M.; McCord, R. A.; Ongaigui, K. C.; Boxer, L. D.; Chang, H. Y.; Chua, K. F. SIRT6 links histone H3 lysine 9 deacetylation to NF-kB-dependent gene expression and organismal life span. Cell 136:62–74; 2009. [118] Kawahara, T. L.; Rapicavoli, N. A.; Wu, A. R.; Qu, K.; Quake, S. R.; Chang, H. Y. Dynamic chromatin localization of Sirt6 shapes stress- and aging-related transcriptional networks. PLoS Genet. 7:e1002153; 2011. [119] Tennen, R. I.; Chua, K. F. Chromatin regulation and genome maintenance by mammalian SIRT6. Trends Biochem. Sci. 36:39–46; 2011. [120] Lombard, D. B.; Alt, F. W.; Cheng, H. L.; Bunkenborg, J.; Streeper, R. S.; Mostoslavsky, R.; Kim, J.; Yancopoulos, G.; Valenzuela, D.; Murphy, A.; Yang, Y.; Chen, Y.; Hirschey, M. D.; Bronson, R. T.; Haigis, M.; Guarente, L. P.; Farese Jr R. V.; Weissman, S.; Verdin, E.; Schwer, B. Mammalian Sir2 homolog SIRT3 regulates global mitochondrial lysine acetylation. Mol. Cell Biol. 27; 2007. 8807-8014. [121] Hirschey, M. D.; Shimazu, T.; Goetzman, E.; Jing, E.; Schwer, B.; Lombard, D. B.; Grueter, C. A.; Harris, C.; Biddinger, S.; Ilkayeva, O. R.; Stevens, R. D.; Li, Y.; Saha, A. K.; Ruderman, N. B.; Bain, J. R.; Newgard, C. B.; Farese Jr R. V.; Alt, F. W.; Kahn, C. R.; Verdin, E. SIRT3 regulates mitochondrial fatty-acid oxidation by reversible enzyme deacetylation. Nature 464:121–125; 2010. [122] Hallows, W. C.; Yu, W.; Smith, B. C.; Devries, M. K.; Ellinger, J. J.; Someya, S.; Shortreed, M. R.; Prolla, T.; Markley, J. L.; Smith, L. M.; Zhao, S.; Guan, K. L.; Denu, J. M. Sirt3 promotes the urea cycle and fatty acid oxidation during dietary restriction. Mol. Cell 41:139–149; 2011. [123] Guarente, L. Mitochondria—a nexus for aging, calorie restriction, and sirtuins? Cell 132:171–176; 2008. [124] Hirschey, M. D.; Shimazu, T.; Jing, E.; Grueter, C. A.; Collins, A. M.; Aouizerat, B.; Stancˇa´kova´, A.; Goetzman, E.; Lam, M. M.; Schwer, B.; Stevens, R. D.; Muehlbauer, M. J.; Kakar, S.; Bass, N. M.; Kuusisto, J.; Laakso, M.; Alt, F. W.; Newgard, C. B.; Farese Jr R. V.; Kahn, C. R.; Verdin, E. SIRT3 deficiency and mitochondrial protein hyperacetylation accelerate the development of the metabolic syndrome. Mol. Cell 44:177–190; 2011. [125] Shimazu, T.; Hirschey, M. D.; Hua, L.; Dittenhafer-Reed, K. E.; Schwer, B.; Lombard, D. B.; Li, Y.; Bunkenborg, J.; Alt, F. W.; Denu, J. M.; Jacobson, M. P.; Verdin, E. SIRT3 deacetylates mitochondrial 3-hydroxy-3-methylglutaryl CoA synthase 2 and regulates ketone body production. Cell Metab. 12:654–661; 2010. [126] Schlicker, C.; Gertz, M.; Papatheodorou, P.; Kachholz, B.; Becker, C. F.; Steegborn, C. Substrates and regulation mechanisms for the human mitochondrial sirtuins Sirt3 and Sirt5. J. Mol. Biol. 382:790–801; 2008. [127] Someya, S.; Yu, W.; Hallows, W. C.; Xu, J.; Vann, J. M.; Leeuwenburgh, C.; Tanokura, M.; Denu, J. M.; Prolla, T. A. Sirt3 mediates reduction of oxidative damage and prevention of age-related hearing loss under caloric restriction. Cell 143:802–812; 2010. [128] Ahn, B. H.; Kim, H. S.; Song, S.; Lee, I. H.; Liu, J.; Vassilopoulos, A.; Deng, C. X.; Finkel, T. A role for the mitochondrial deacetylase Sirt3 in regulating energy homeostasis. Proc. Natl. Acad. Sci. USA 105:14447–14452; 2008. [129] Finley, L. W.; Haas, W.; Desquiret-Dumas, V.; Wallace, D. C.; Procaccio, V.; Gygi, S. P.; Haigis, M. C. Succinate dehydrogenase is a direct target of sirtuin 3 deacetylase activity. PLoS One 6:e23295; 2011. [130] Jing, E.; Emanuelli, B.; Hirschey, M. D.; Boucher, J.; Lee, K. Y.; Lombard, D.; Verdin, E. M.; Kahn, C. R. Sirtuin-3 (Sirt3) regulates skeletal muscle metabolism and insulin signaling via altered mitochondrial oxidation and reactive oxygen species production. Proc. Natl. Acad. Sci. USA 108:14608–14613; 2011. [131] Qiu, X.; Brown, K.; Hirschey, M. D.; Verdin, E.; Chen, D. Calorie restriction reduces oxidative stress by SIRT3-mediated SOD2 activation. Cell Metab. 12:662–667; 2010. [132] Tao, R.; Coleman, M. C.; Pennington, J. D.; Ozden, O.; Park, S. H.; Jiang, H.; Kim, H. S.; Flynn, C. R.; Hill, S.; Hayes McDonald, W.; Olivier, A. K.; Spitz, D. R.; Gius, D. Sirt3-mediated deacetylation of evolutionarily conserved lysine 122 regulates MnSOD activity in response to stress. Mol. Cell 40:893–904; 2010. [133] Chen, Y.; Zhang, J.; Lin, Y.; Lei, Q.; Guan, K. L.; Zhao, S.; Xiong, Y. Tumour suppressor SIRT3 deacetylates and activates manganese superoxide dismutase to scavenge ROS. EMBO Rep 12:534–541; 2011. [134] Sundaresan, N. R.; Gupta, M.; Kim, G.; Rajamohan, S. B.; Isbatan, A.; Gupta, M. P. Sirt3 blocks the cardiac hypertrophic response by augmenting Foxo3adependent antioxidant defense mechanisms in mice. J. Clin. Invest. 119:2758–2771; 2009. [135] Kim, H. S.; Patel, K.; Muldoon-Jacobs, K.; Bisht, K. S.; Aykin-Burns, N.; Pennington, J. D.; van der Meer, R.; Nguyen, P.; Savage, J.; Owens, K. M.; Vassilopoulos, A.; Ozden, O.; Park, S. H.; Singh, K. K.; Abdulkadir, S. A.; Spitz, D. R.; Deng, C. X.; Gius, D. SIRT3 is a mitochondria-localized tumor suppressor required for maintenance of mitochondrial integrity and metabolism during stress. Cancer Cell 17:41–52; 2010.

161

[136] Pellegrini, L.; Pucci, B.; Villanova, L.; Marino, M. L.; Marfe, G.; Sansone, L.; Vernucci, E.; Bellizzi, D.; Reali, V.; Fini, M.; Russo, M. A.; Tafani, M. SIRT3 protects from hypoxia and staurosporine-mediated cell death by maintaining mitochondrial membrane potential and intracellular pH. Cell Death Differ. 19:1815–1825; 2012. [137] Zhang, B.; Cui, S.; Bai, X.; Zhuo, L.; Hong, Q.; Fu, B.; Wang, J.; Chen, X.; Cai, G.SIRT3 overexpression antagonizes high glucose accelerated cellular senescence in human diploid fibroblasts via the SIRT3-FOXO1 signaling pathway. Submitted for publication. [138] Finley, L. W.; Carracedo, A.; Lee, J.; Souza, A.; Egia, A.; Zhang, J.; TeruyaFeldstein, J.; Moreira, P. I.; Cardoso, S. M.; Clish, C. B.; Pandolfi, P. P.; Haigis, M. C. SIRT3 opposes reprogramming of cancer cell metabolism through HIF1a destabilization. Cancer Cell 19:416–428; 2011. [139] Li, D.; Dammer, E. B.; Sewer, M. B. Resveratrol stimulates cortisol biosynthesis by activating SIRT-dependent deacetylation of P450scc. Endocrinology 153:3258–3268; 2012. [140] Nasrin, N.; Wu, X.; Fortier, E.; Feng, Y.; Bare, O. C.; Chen, S.; Ren, X.; Wu, Z.; Streeper, R. S.; Bordone, L. SIRT4 regulates fatty acid oxidation and mitochondrial gene expression in liver and muscle cells. J. Biol. Chem. 285:31995–32002; 2010. [141] Ogura, M.; Nakamura, Y.; Tanaka, D.; Zhuang, X.; Fujita, Y.; Obara, A.; Hamasaki, A.; Hosokawa, M.; Inagaki, N. Overexpression of SIRT5 confirms its involvement in deacetylation and activation of carbamoyl phosphate synthetase 1. Biochem. Biophys. Res. Commun. 393:73–78; 2010. [142] Wang, F.; Tong, Q. SIRT2 suppresses adipocyte differentiation by deacetylating FOXO1 and enhancing FOXO1’s repressive interaction with PPARg. Mol. Biol. Cell 20:801–808; 2009. [143] Wang, F.; Nguyen, M.; Qin, F. X.; Tong, Q. SIRT2 deacetylates FOXO3a in response to oxidative stress and caloric restriction. Aging Cell 6:505–514; 2007. [144] Wang, F.; Chan, C. H.; Chen, K.; Guan, X.; Lin, H. K.; Tong, Q. Deacetylation of FOXO3 by SIRT1 or SIRT2 leads to Skp2-mediated FOXO3 ubiquitination and degradation. Oncogene 31:1546–1557; 2012. [145] Li, W.; Zhang, B.; Tang, J.; Cao, Q.; Wu, Y.; Wu, C.; Guo, J.; Ling, E. A.; Liang, F. Sirtuin 2, a mammalian homolog of yeast silent information regulator-2 longevity regulator, is an oligodendroglial protein that decelerates cell differentiation through deacetylating a-tubulin. J. Neurosci. 27:2606–2616; 2007. [146] Inoue, T.; Hiratsuka, M.; Osaki, M.; Oshimura, M. The molecular biology of mammalian SIRT proteins: SIRT2 in cell cycle regulation. Cell Cycle 6:1011–1108; 2007. [147] Das, C.; Lucia, M. S.; Hansen, K. C.; Tyler, J. K. CBP/p300-mediated acetylation of histone H3 on lysine 56. Nature 459:113–117; 2009. [148] de Oliveira, R. M.; Sarkander, J.; Kazantsev, A. G.; Outeiro, T. F. SIRT2 as a therapeutic target for age-related disorders. Front. Pharmacol. 3(article 82):1–9; 2012. [149] Black, J. C.; Mosley, A.; Kitada, T.; Washburn, M.; Carey, M. The SIRT2 deacetylase regulates autoacetylation of p300. Mol. Cell 32:449–455; 2008. [150] Han, Y.; Jin, Y. H.; Kim, Y. J.; Kang, B. Y.; Choi, H. J.; Kim, D. W.; Yeo, C. Y.; Lee, K. Y. Acetylation of Sirt2 by p300 attenuates its deacetylase activity. Biochem. Biophys. Res. Commun. 375:576–580; 2008. [151] Jin, Y. H.; Kim, Y. J.; Kim, D. W.; Baek, K. H.; Kang, B. Y.; Yeo, C. Y.; Lee, K. Y. Sirt2 interacts with 14-3-3 b/g and down-regulates the activity of p53. Biochem. Biophys. Res. Commun 368:690–695; 2008. [152] Fu, H.; Subramanian, R. R.; Masters, S. C. 14-3-3 proteins: structure, function, and regulation. Annu. Rev. Pharmacol. Toxicol. 40:617–647; 2000. [153] Bae, N. S.; Swanson, M. J.; Vassilev, A.; Howard, B. H. Human histone deacetylase SIRT2 interacts with the homeobox transcription factor HOXA10. J. Biochem. 135:695–700; 2004. ¨ [154] Rothgiesser, K. M.; Erener, S.; Waibel, S.; Luscher, B.; Hottiger, M. O. SIRT2 regulates NF-kB dependent gene expression through deacetylation of p65 Lys310. J. Cell Sci. 123:4251–4258; 2010. [155] Jiang, W.; Wang, S.; Xiao, M.; Lin, Y.; Zhou, L.; Lei, Q.; Xiong, Y.; Guan, K. L.; Zhao, S. Acetylation regulates gluconeogenesis by promoting PEPCK1 degradation via recruiting the UBR5 ubiquitin ligase. Mol. Cell 43:33–44; 2011. [156] Beirowski, B.; Gustin, J.; Armour, S. M.; Yamamoto, H.; Viader, A.; North, B. J.; Micha´n, S.; Baloh, R. H.; Golden, J. P.; Schmidt, R. E.; Sinclair, D. A.; Auwerx, J.; Milbrandt, J. Sir-two-homolog 2 (Sirt2) modulates peripheral myelination through polarity protein Par-3/atypical protein kinase C (aPKC) signaling. Proc. Natl. Acad. Sci. USA 108:E952–E961; 2011. [157] Ford, E.; Voit, R.; Liszt, G.; Magin, C.; Grummt, I.; Guarente, L. Mammalian Sir2 homolog SIRT7 is an activator of RNA polymerase I transcription. Genes Dev. 20:1075–1080; 2006. [158] Tsai, Y. C.; Greco, T. M.; Boonmee, A.; Miteva, Y.; Cristea, I. M. Functional proteomics establishes the interaction of SIRT7 with chromatin remodeling complexes and expands Its role in regulation of RNA polymerase I transcription. Mol. Cell. Proteomics 11:60–76; 2012. [159] Nemoto, S.; Fergusson, M. M.; Finkel, T. Nutrient availability regulates SIRT1 through a forkhead-dependent pathway. Science 306:2105–2108; 2004. [160] Coste, A.; Louet, J. F.; Lagouge, M.; Lerin, C.; Antal, M. C.; Meziane, H.; Schoonjans, K.; Puigserver, P.; O’Malley, B. W.; Auwerx, J. The genetic ablation of SRC-3 protects against obesity and improves insulin sensitivity by reducing the acetylation of PGC-1a. Proc. Natl. Acad. Sci. USA 105:17187–17192; 2008. [161] Hayashida, S.; Arimoto, A.; Kuramoto, Y.; Kozako, T.; Honda, S.; Shimeno, H.; Soeda, S. Fasting promotes the expression of SIRT1, an NAD þ -dependent

162

[162]

[163]

[164]

[165]

[166]

[167]

[168]

[169]

[170] [171] [172] [173]

[174] [175]

[176]

[177]

[178]

[179]

[180]

[181]

[182]

[183] [184] [185]

[186]

[187] [188] [189]

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

protein deacetylase, via activation of PPARa in mice. Mol. Cell. Biochem. 339:285–292; 2010. Okazaki, M.; Iwasaki, Y.; Nishiyama, M.; Taguchi, T.; Tsugita, M.; Nakayama, S.; Kambayashi, M.; Hashimoto, K.; Terada, Y. PPARb/d regulates the human SIRT1 gene transcription via Sp1. Endocr. J. 57:403–413; 2010. Noriega, L. G.; Feige, J. N.; Canto, C.; Yamamoto, H.; Yu, J.; Herman, M. A.; Mataki, C.; Kahn, B. B.; Auwerx, J. CREB and ChREBP oppositely regulate SIRT1 expression in response to energy availability. EMBO Rep 12:1069–1076; 2011. Wang, C.; Chen, L.; Hou, X.; Li, Z.; Kabra, N.; Ma, Y.; Nemoto, S.; Finkel, T.; Gu, W.; Cress, W. D.; Chen, J. Interactions between E2F1 and SirT1 regulate apoptotic response to DNA damage. Nat. Cell Biol. 8:1025–1031; 2006. Han, L.; Zhou, R.; Niu, J.; McNutt, M. A.; Wang, P.; Tong, T. SIRT1 is regulated by a PPARg-SIRT1 negative feedback loop associated with senescence. Nucleic Acids Res. 38:7458–7471; 2010. Chen, W. Y.; Wang, D. H.; Yen, R. C.; Luo, J.; Gu, W.; Baylin, S. B. Tumor suppressor HIC1 directly regulates SIRT1 to modulate p53-dependent DNAdamage responses. Cell 123:437–448; 2005. Zhang, Q.; Wang, S. Y.; Fleuriel, C.; Leprince, D.; Rocheleau, J. V.; Piston, D. W.; Goodman, R. H. Metabolic regulation of SIRT1 transcription via a HIC1:CtBP corepressor complex. Proc. Natl. Acad. Sci. USA 104:829–833; 2007. Giralt, A.; Hondares, E.; Villena, J. A.; Ribas, F.; Dı´az-Delfı´n, J.; Giralt, M.; Iglesias, R.; Villarroya, F. Peroxisome proliferator-activated receptor-gamma coactivator-1a controls transcription of the Sirt3 gene, an essential component of the thermogenic brown adipocyte phenotype. J. Biol. Chem. 286:16958–16966; 2011. Abdelmohsen, K.; Pullmann Jr R.; Lal, A.; Kim, H. H.; Galban, S.; Yang, X.; Blethrow, J. D.; Walker, M.; Shubert, J.; Gillespie, D. A.; Furneaux, H.; Gorospe, M. Phosphorylation of HuR by Chk2 regulates SIRT1 expression. Mol. Cell 25:543–557; 2007. Bartel, D. P. MicroRNAs: target recognition and regulatory functions. Cell 136:215–233; 2009. Yamakuchi, M. MicroRNA regulation of SIRT1. Front. Physiol. 3(article 68):1–8; 2012. Yamakuchi, M.; Ferlito, M.; Lowenstein, C. J. miR-34a repression of SIRT1 regulates apoptosis. Proc. Natl. Acad. Sci. USA 105:13421–13426; 2008. Rane, S.; He, M.; Sayed, D.; Vashistha, H.; Malhotra, A.; Sadoshima, J.; Vatner, D. E.; Vatner, S. F.; Abdellatif, M. Downregulation of miR-199a derepresses hypoxia-inducible factor-1a and Sirtuin 1 and recapitulates hypoxia preconditioning in cardiac myocytes. Circ. Res. 104:879–886; 2009. ¨ Flick, F.; Luscher, B. Regulation of sirtuin function by posttranslational modifications. Front.Pharmacol. 3(article 29):1–13; 2012. Sasaki, T.; Maier, B.; Koclega, K. D.; Chruszcz, M.; Gluba, W.; Stukenberg, P. T.; Minor, W.; Scrable, H. Phosphorylation regulates SIRT1 function. PLoS One 3:e4020; 2008 13 pp. Nasrin, N.; Kaushik, V. K.; Fortier, E.; Wall, D.; Pearson, K. J.; de Cabo, R.; Bordone, L. JNK1 phosphorylates SIRT1 and promotes its enzymatic activity. PLoS One 4:e8414; 2009. ¨ un, ¨ C. Z.; Uysal, K. T.; Maeda, K.; Hirosumi, J.; Tuncman, G.; Chang, L.; Gorg Karin, M.; Hotamisligil, G. S. A central role for JNK in obesity and insulin resistance. Nature 420:333–336; 2002. Guo, X.; Williams, J. G.; Schug, T. T.; Li, X. DYRK1A and DYRK3 promote cell survival through phosphorylation and activation of SIRT1. J. Biol. Chem. 285:13223–13232; 2010. Lin, Z.; Yang, H.; Kong, Q.; Li, J.; Lee, S. M.; Gao, B.; Dong, H.; Wei, J.; Song, J.; Zhang, D. D.; Fang, D. USP22 antagonizes p53 transcriptional activation by deubiquitinating Sirt1 to suppress cell apoptosis and is required for mouse embryonic development. Mol. Cell 46:484–494; 2012. ¨ Pandithage, R.; Lilischkis, R.; Harting, K.; Wolf, A.; Jedamzik, B.; Luscher¨ B.; Luscher, ¨ Firzlaff, J.; Vervoorts, J.; Lasonder, E.; Kremmer, E.; Knoll, B. The regulation of SIRT2 function by cyclin-dependent kinases affects cell motility. J. Cell Biol. 180:915–929; 2008. Yang, Y.; Fu, W.; Chen, J.; Olashaw, N.; Zhang, X.; Nicosia, S. V.; Bhalla, K.; Bai, W. SIRT1 sumoylation regulates its deacetylase activity and cellular response to genotoxic stress. Nat. Cell Biol. 9:1253–1262; 2007. Kim, E. J.; Kho, J. H.; Kang, M. R.; Um, S. J. Active regulator of SIRT1 cooperates with SIRT1 and facilitates suppression of p53 activity. Mol. Cell 28:277–290; 2007. Zhao, W.; Kruse, J. P.; Tang, Y.; Jung, S. Y.; Qin, J.; Gu, W. Negative regulation of the deacetylase SIRT1 by DBC1. Nature 451:587–590; 2008. Kim, J. E.; Chen, J.; Lou, Z. DBC1 is a negative regulator of SIRT1. Nature 451:583–586; 2008. Escande, C.; Chini, C. C.; Nin, V.; Dykhouse, K. M.; Novak, C. M.; Levine, J.; van Deursen, J.; Gores, G. J.; Chen, J.; Lou, Z.; Chini, E. N. Deleted in breast cancer-1 regulates SIRT1 activity and contributes to high-fat diet-induced liver steatosis in mice. J. Clin. Invest. 120:545–558; 2010. Wang, R. H.; Li, C.; Deng, C. X. Liver steatosis and increased ChREBP expression in mice carrying a liver specific SIRT1 null mutation under a normal feeding condition. Int. J. Biol. Sci. 6:682–690; 2010. Li, Z.; Chen, L.; Kabra, N.; Wang, C.; Fang, J.; Chen, J. Inhibition of SUV39H1 methyltransferase activity by DBC1. J. Biol. Chem. 284:10361–10366; 2009. Yuan, J.; Luo, K.; Liu, T.; Lou, Z. Regulation of SIRT1 activity by genotoxic stress. Genes Dev. 26:791–796; 2012. Yu, E. J.; Kim, S. H.; Heo, K.; Ou, C. Y.; Stallcup, M. R.; Kim, J. H. Reciprocal roles of DBC1 and SIRT1 in regulating estrogen receptor a activity and coactivator synergy. Nucleic Acids Res. 39:6932–6943; 2011.

[190] Mulligan, P.; Yang, F.; Di Stefano, L.; Ji, J. Y.; Ouyang, J.; Nishikawa, J. L.; Toiber, D.; Kulkarni, M.; Wang, Q.; Najafi-Shoushtari, S. H.; Mostoslavsky, R.; ¨ A. M. A SIRT1-LSD1 corepressor Gygi, S. P.; Gill, G.; Dyson, N. J.; Na¨ ar, complex regulates Notch target gene expression and development. Mol. Cell 42:689–699; 2011. [191] Kuzmichev, A.; Margueron, R.; Vaquero, A.; Preissner, T. S.; Scher, M.; Kirmizis, A.; Ouyang, X.; Brockdorff, N.; Abate-Shen, C.; Farnham, P.; Reinberg, D. Composition and histone substrates of polycomb repressive group complexes change during cellular differentiation. Proc. Natl. Acad. Sci. USA 102:1859–1864; 2005. [192] Houtkooper, R. H.; Pirinen, E.; Auwerx, J. Sirtuins as regulators of metabolism and healthspan. Nat. Rev. Mol. Cell. Biol. 13:225–238; 2012. [193] Rodgers, J. T.; Puigserver, P. Fasting-dependent glucose and lipid metabolic response through hepatic sirtuin 1. Proc. Natl. Acad. Sci. USA 104:12861–12866; 2007. [194] Wang, R. H.; Kim, H. S.; Xiao, C.; Xu, X.; Gavrilova, O.; Deng, C. X. Hepatic Sirt1 deficiency in mice impairs mTorc2/Akt signaling and results in hyperglycemia, oxidative damage, and insulin resistance. J. Clin. Invest. 121:4477–4490; 2011. [195] Herzig, S.; Long, F.; Jhala, U. S.; Hedrick, S.; Quinn, R.; Bauer, A.; Rudolph, D.; Schutz, G.; Yoon, C.; Puigserver, P.; Spiegelman, B.; Montminy, M. CREB regulates hepatic gluconeogenesis through the coactivator PGC-1. Nature 413:179–183; 2001. [196] Yoon, J. C.; Puigserver, P.; Chen, G.; Donovan, J.; Wu, Z.; Rhee, J.; Adelmant, G.; Stafford, J.; Kahn, C. R.; Granner, D. K.; Newgard, C. B.; Spiegelman, B. M. Control of hepatic gluconeogenesis through the transcriptional coactivator PGC-1. Nature 413:131–138; 2001. [197] Herzog, B.; Hall, R. K.; Wang, X. L.; Waltner-Law, M.; Granner, D. K. Peroxisome proliferator-activated receptor gamma coactivator-1a, as a transcription amplifier, is not essential for basal and hormone-induced phosphoenolpyruvate carboxykinase gene expression. Mol. Endocrinol. 18:807–819; 2004. [198] Canto´, C.; Auwerx, J. AMP-activated protein kinase and its downstream transcriptional pathways. Cell. Mol. Life Sci. 67:3407–3423; 2010. [199] Buler, M.; Aatsinki, S. M.; Skoumal, R.; Hakkola, J. Energy sensing factors PGC-1a and SIRT1 modulate PXR expression and function. Biochem. Pharmacol. 82:2008–2015; 2011. [200] Yang, S. J.; Choi, J. M.; Chae, S. W.; Kim, W. J.; Park, S. E.; Rhee, E. J.; Lee, W. Y.; Oh, K. W.; Park, S. W.; Kim, S. W.; Park, C. Y. Activation of peroxisome proliferator-activated receptor gamma by rosiglitazone increases sirt6 expression and ameliorates hepatic steatosis in rats. PLoS One 6:e17057; 2011. [201] Bordone, L.; Motta, M. C.; Picard, F.; Robinson, A.; Jhala, U. S.; Apfeld, J.; McDonagh, T.; Lemieux, M.; McBurney, M.; Szilvasi, A.; Easlon, E. J.; Lin, S. J.; Guarente, L. Sirt1 regulates insulin secretion by repressing UCP2 in pancreatic beta cells. PLoS Biol. 4:e31; 2006. [202] Moynihan, K. A.; Grimm, A. A.; Plueger, M. M.; Bernal-Mizrachi, E.; Ford, E.; Cras-Me´neur, C.; Permutt, M. A.; Imai, S. Increased dosage of mammalian Sir2 in pancreatic beta cells enhances glucose-stimulated insulin secretion in mice. Cell Metab. 2:105–117; 2005. [203] Ramsey, K. M.; Mills, K. F.; Satoh, A.; Imai, S. Age-associated loss of Sirt1mediated enhancement of glucose-stimulated insulin secretion in beta cellspecific Sirt1-overexpressing (BESTO) mice. Aging Cell 7:78–88; 2008. [204] Braidy, N.; Guillemin, G. J.; Mansour, H.; Chan-Ling, T.; Poljak, A.; Grant, R. Age related changes in NAD þ metabolism oxidative stress and Sirt1 activity in Wistar rats. PLoS One 6:e19194; 2011. [205] Zhang, C. Y.; Baffy, G.; Perret, P.; Krauss, S.; Peroni, O.; Grujic, D.; Hagen, T.; Vidal-Puig, A. J.; Boss, O.; Kim, Y. B.; Zheng, X. X.; Wheeler, M. B.; Shulman, G. I.; Chan, C. B.; Lowell, B. B. Uncoupling protein-2 negatively regulates insulin secretion and is a major link between obesity, b cell dysfunction, and type 2 diabetes. Cell 105:745–755; 2001. [206] Ramachandran, D.; Roy, U.; Garg, S.; Ghosh, S.; Pathak, S.; KolthurSeetharam, U. Sirt1 and mir-9 expression is regulated during glucosestimulated insulin secretion in pancreatic b-islets. FEBS J 278:1167–1174; 2011. [207] Bastien-Dionne, P. O.; Valenti, L.; Kon, N.; Gu, W.; Buteau, J. Glucagon-like peptide 1 inhibits the sirtuin deacetylase SirT1 to stimulate pancreatic b-cell mass expansion. Diabetes 60:3217–3222; 2011. [208] Ahuja, N.; Schwer, B.; Carobbio, S.; Waltregny, D.; North, B. J.; Castronovo, V.; Maechler, P.; Verdin, E. Regulation of insulin secretion by SIRT4, a mitochondrial ADP-ribosyltransferase. J. Biol. Chem. 282:33583–33592; 2007. [209] Gerhart-Hines, Z.; Rodgers, J. T.; Bare, O.; Lerin, C.; Kim, S. H.; Mostoslavsky, R.; Alt, F. W.; Wu, Z.; Puigserver, P. Metabolic control of muscle mitochondrial function and fatty acid oxidation through SIRT1/PGC-1a. EMBO J. 26:1913–1923; 2007. [210] Philp, A.; Chen, A.; Lan, D.; Meyer, G. A.; Murphy, A. N.; Knapp, A. E.; Olfert, I. M.; McCurdy, C. E.; Marcotte, G. R.; Hogan, M. C.; Baar, K.; Schenk, S. Sirtuin 1 (SIRT1) deacetylase activity is not required for mitochondrial biogenesis or peroxisome proliferator-activated receptor-g coactivator-1a (PGC-1a) deacetylation following endurance exercise. J. Biol. Chem. 286:30561–30570; 2011. [211] Gurd, B. J.; Yoshida, Y.; McFarlan, J. T.; Holloway, G. P.; Moyes, C. D.; Heigenhauser, G. J.; Spriet, L.; Bonen, A. Nuclear SIRT1 activity, but not protein content, regulates mitochondrial biogenesis in rat and human skeletal muscle. Am. J. Physiol. Regul. Integr. Comp. Physiol 301:R67–R75; 2011.

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

[212] Satoh, A.; Brace, C. S.; Ben-Josef, G.; West, T.; Wozniak, D. F.; Holtzman, D. M.; Herzog, E. D.; Imai, S. SIRT1 promotes the central adaptive response to diet restriction through activation of the dorsomedial and lateral nuclei of the hypothalamus. J. Neurosci. 30:10220–10232; 2010. [213] Schenk, S.; McCurdy, C. E.; Philp, A.; Chen, M. Z.; Holliday, M. J.; Bandyopadhyay, G. K.; Osborn, O.; Baar, K.; Olefsky, J. M. Sirt1 enhances skeletal muscle insulin sensitivity in mice during caloric restriction. J. Clin. Invest. 121:4281–4288; 2011. ¨ o, ¨ S.; Durand, C.; Molin, L.; Carey, A. L.; El-Osta, A.; Kingwell, B. A.; [214] Frojd Febbraio, M. A.; Solari, F.; Vidal, H.; Pirola, L. Phosphoinositide 3-kinase as a novel functional target for the regulation of the insulin signaling pathway by SIRT1. Mol. Cell. Endocrinol. 335:166–176; 2011. [215] Liu, C. T.; Brooks, G. A. Mild heat stress induces mitochondrial biogenesis in C2C12 myotubes. J. Appl. Physiol. 112:354–361; 2012. [216] Alamdari, N.; Aversa, Z.; Castillero, E.; Gurav, A.; Petkova, V.; Tizio, S.; Hasselgren, P. O. Resveratrol prevents dexamethasone-induced expression of the muscle atrophy-related ubiquitin ligases atrogin-1 and MuRF1 in cultured myotubes through a SIRT1-dependent mechanism. Biochem. Biophys. Res. Commun. 417:528–533; 2012. [217] Jang, Y. C.; Liu, Y.; Hayworth, C. R.; Bhattacharya, A.; Lustgarten, M. S.; Muller, F. L.; Chaudhuri, A.; Qi, W.; Li, Y.; Huang, J. Y.; Verdin, E.; Richardson, A.; Van Remmen, H. Dietary restriction attenuates age-associated muscle atrophy by lowering oxidative stress in mice even in complete absence of CuZnSOD. Aging Cell 11:770–782; 2012. [218] Palacios, O. M.; Carmona, J. J.; Michan, S.; Chen, K. Y.; Manabe, Y.; Ward 3rd J. L.; Goodyear, L. J.; Tong, Q. Diet and exercise signals regulate SIRT3 and activate AMPK and PGC-1alpha in skeletal muscle. Aging (Albany NY) 1:771–783; 2009. [219] Fernandez-Marcos, P. J.; Jeninga, E. H.; Canto, C.; Harach, T.; de Boer, V. C.; Andreux, P.; Moullan, N.; Pirinen, E.; Yamamoto, H.; Houten, S. M.; Schoonjans, K.; Auwerx, J. Muscle or liver-specific Sirt3 deficiency induces hyperacetylation of mitochondrial proteins without affecting global metabolic homeostasis. Sci. Rep 2:425; 2012. [220] Gurd, B. J.; Holloway, G. P.; Yoshida, Y.; Bonen, A. In mammalian muscle, SIRT3 is present in mitochondria and not in the nucleus; and SIRT3 is upregulated by chronic muscle contraction in an adenosine monophosphate-activated protein kinase-independent manner. Metabolism 61:733–741; 2012. [221] Li, Y.; Xu, W.; McBurney, M. W.; Longo, V. D. SirT1 inhibition reduces IGF-I/IRS2/Ras/ERK1/2 signaling and protects neurons. Cell Metab. 8:38–48; 2008. [222] Cohen, D. E.; Supinski, A. M.; Bonkowski, M. S.; Donmez, G.; Guarente, L. P. Neuronal SIRT1 regulates endocrine and behavioral responses to calorie restriction. Genes Dev. 23:2812–2817; 2009. [223] Geng, Y. Q.; Li, T. T.; Liu, X. Y.; Li, Z. H.; Fu, Y. C. SIRT1 and SIRT5 activity expression and behavioral responses to calorie restriction. J. Cell. Biochem. 112:3755–3761; 2011. [224] Libert, S.; Pointer, K.; Bell, E. L.; Das, A.; Cohen, D. E.; Asara, J. M.; Kapur, K.; Bergmann, S.; Preisig, M.; Otowa, T.; Kendler, K. S.; Chen, X.; Hettema, J. M.; van den Oord, E. J.; Rubio, J. P.; Guarente, L. SIRT1 activates MAO-A in the brain to mediate anxiety and exploratory drive. Cell 147:1459–1472; 2011. [225] Fusco, S.; Ripoli, C.; Podda, M. V.; Ranieri, S. C.; Leone, L.; Toietta, G.; ¨ McBurney, M. W.; Schutz, G.; Riccio, A.; Grassi, C.; Galeotti, T.; Pani, G. A role for neuronal cAMP responsive-element binding (CREB)-1 in brain responses to calorie restriction. Proc. Natl. Acad. Sci. USA 109:621–626; 2012. [226] Torres, G.; Dileo, J. N.; Hallas, B. H.; Horowitz, J. M.; Leheste, J. R. Silent information regulator 1 mediates hippocampal plasticity through presenilin1. Neuroscience 179:32–40; 2011. [227] Zocchi, L.; Sassone-Corsi, P. SIRT1-mediated deacetylation of MeCP2 contributes to BDNF expression. Epigenetics 7:695–700; 2012. [228] Bayod, S.; Del Valle, J.; Canudas, A. M.; Lalanza, J. F.; Sanchez-Roige, S.; Camins, A.; Escorihuela, R. M.; Palla s, M. Long-term treadmill exercise induces neuroprotective molecular changes in rat brain. J. Appl. Physiol. 111:1380–1390; 2011. [229] Coppari, R. Metabolic actions of hypothalamic SIRT1. Trends Endocrinol. Metab. 23:179–185; 2012. [230] Hasegawa, K.; Kawahara, T.; Fujiwara, K.; Shimpuku, M.; Sasaki, T.; Kitamura, T.; Yoshikawa, K. Necdin controls Foxo1 acetylation in hypothalamic arcuate neurons to modulate the thyroid axis. J. Neurosci. 32:5562–5572; 2012. [231] Zhu, H.; Zhao, L.; Wang, E.; Dimova, N.; Liu, G.; Feng, Y.; Cambi, F. The QKIPLP pathway controls SIRT2 abundance in CNS myelin. Glia 60:69–82; 2012. [232] Werner, H. B.; Kuhlmann, K.; Shen, S.; Uecker, M.; Schardt, A.; Dimova, K.; ¨ Orfaniotou, F.; Dhaunchak, A.; Brinkmann, B. G.; Mobius, W.; Guarente, L.; Casaccia-Bonnefil, P.; Jahn, O.; Nave, K. A. Proteolipid protein is required for transport of sirtuin 2 into CNS myelin. J. Neurosci. 27:7717–7730; 2007. [233] Ji, S.; Doucette, J. R.; Nazarali, A. J. Sirt2 is a novel in vivo downstream target of Nkx2.2 and enhances oligodendroglial cell differentiation. J. Mol. Cell. Biol. 3:351–359; 2012. [234] Southwood, C. M.; Peppi, M.; Dryden, S.; Tainsky, M. A.; Gow, A. Microtubule deacetylases, SirT2 and HDAC6, in the nervous system. Neurochem. Res. 32:187–195; 2007. [235] Harting, K.; Kn¨oll, B. SIRT2-mediated protein deacetylation: an emerging key regulator in brain physiology and pathology. Eur. J. Cell Biol. 89:262–269; 2010. [236] Renault, V. M.; Rafalski, V. A.; Morgan, A. A.; Salih, D. A.; Brett, J. O.; Webb, A. E.; Villeda, S. A.; Thekkat, P. U.; Guillerey, C.; Denko, N. C.; Palmer, T. D.; Butte, A. J.; Brunet, A. FoxO3 regulates neural stem cell homeostasis. Cell Stem Cell 5:527–539; 2009.

163

[237] Kim, M. J.; Ahn, K.; Park, S. H.; Kang, H. J.; Jang, B. G.; Oh, S. J.; Oh, S. M.; Jeong, Y. J.; Heo, J. I.; Suh, J. G.; Lim, S. S.; Ko, Y. J.; Huh, S. O.; Kim, S. C.; Park, J. B.; Kim, J.; Kim, J. I.; Jo, S. A.; Lee, J. Y. SIRT1 regulates tyrosine hydroxylase expression and differentiation of neuroblastoma cells via FOXO3a. FEBS Lett 583:1183–1188; 2009. [238] Maxwell, M. M.; Tomkinson, E. M.; Nobles, J.; Wizeman, J. W.; Amore, A. M.; Quinti, L.; Chopra, V.; Hersch, S. M.; Kazantsev, A. G. The Sirtuin 2 microtubule deacetylase is an abundant neuronal protein that accumulates in the aging CNS. Hum. Mol. Genet 20:3986–3996; 2011. [239] Kim, S. H.; Lu, H. F.; Alano, C. C. Neuronal Sirt3 protects against excitotoxic injury in mouse cortical neuron culture. PLoS One 6:e14731; 2011. [240] Bordone, L.; Cohen, D.; Robinson, A.; Motta, M. C.; van Veen, E.; Czopik, A.; Steele, A. D.; Crowe, H.; Marmor, S.; Luo, J.; Gu, W.; Guarente, L. SIRT1 transgenic mice show phenotypes resembling calorie restriction. Aging Cell 6:759–767; 2007. [241] Li, Y.; Xu, S.; Giles, A.; Nakamura, K.; Lee, J. W.; Hou, X.; Donmez, G.; Li, J.; Luo, Z.; Walsh, K.; Guarente, L.; Zang, M. Hepatic overexpression of SIRT1 in mice attenuates endoplasmic reticulum stress and insulin resistance in the liver. FASEB J 25:1664–1679; 2011. [242] Banks, A. S.; Kon, N.; Knight, C.; Matsumoto, M.; Gutie´rrez-Jua´rez, R.; Rossetti, L.; Gu, W.; Accili, D. SirT1 gain of function increases energy efficiency and prevents diabetes in mice. Cell Metab. 8:333–341; 2008. ¨ M. H. [243] Pfluger, P. T.; Herranz, D.; Velasco-Miguel, S.; Serrano, M.; Tschop, Sirt1 protects against high-fat diet-induced metabolic damage. Proc. Natl. Acad. Sci. USA 105:9793–9798; 2008. [244] Lagouge, M.; Argmann, C.; Gerhart-Hines, Z.; Meziane, H.; Lerin, C.; Daussin, F.; Messadeq, N.; Milne, J.; Lambert, P.; Elliott, P.; Geny, B.; Laakso, M.; Puigserver, P.; Auwerx, J. Resveratrol Improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1a. Cell 127:1109–1122; 2006. [245] Xiong, S.; Salazar, G.; Patrushev, N.; Alexander, R. W. FoxO1 mediates an autofeedback loop regulating SIRT1 expression. J. Biol. Chem. 286:5289–5299; 2011. [246] Suwa, M.; Nakano, H.; Radak, Z.; Kumagai, S. Short-term adenosine monophosphate-activated protein kinase activator 5-aminoimidazole-4carboxamide-1-b-D-ribofuranoside treatment increases the sirtuin 1 protein expression in skeletal muscle. Metabolism 60:394–403; 2011. [247] Li, P.; Zhao, Y.; Wu, X.; Xia, M.; Fang, M.; Iwasaki, Y.; Sha, J.; Chen, Q.; Xu, Y.; Shen, A. Interferon gamma (IFN-g) disrupts energy expenditure and metabolic homeostasis by suppressing SIRT1 transcription. Nucleic Acids Res. 40:1609–1620; 2012. [248] Costa Cdos, S.; Hammes, T. O.; Rohden, F.; Margis, R.; Bortolotto, J. W.; Padoin, A. V.; Mottin, C. C.; Guaragna, R. M. SIRT1 transcription is decreased in visceral adipose tissue of morbidly obese patients with severe hepatic steatosis. Obes. Surg. 20:633–639; 2010. [249] Bai, P.; Houten, S. M.; Huber, A.; Schreiber, V.; Watanabe, M.; Kiss, B.; de Murcia, G.; Auwerx, J.; Me´nissier-de Murcia, J. Poly(ADP-ribose) polymerase-2 controls adipocyte differentiation and adipose tissue function through the regulation of the activity of the retinoid X receptor/peroxisome proliferatoractivated receptor-g heterodimer. J. Biol. Chem. 282:37738–37746; 2007. [250] Chalkiadaki, A.; Guarente, L. Metabolic signals regulate SIRT1 expression. EMBO Rep 12:985–986; 2011. [251] Uyeda, K.; Repa, J. J. Carbohydrate response element binding protein, ChREBP, a transcription factor coupling hepatic glucose utilization and lipid synthesis. Cell Metab 4:107–110; 2006. [252] Bell, E. L.; Guarente, L. The SirT3 divining rod points to oxidative stress. Mol. Cell 42:561–568; 2011. [253] Lee, J.; Padhye, A.; Sharma, A.; Song, G.; Miao, J.; Mo, Y. Y.; Wang, L.; Kemper, J. K. A pathway involving farnesoid X receptor and small heterodimer partner positively regulates hepatic sirtuin 1 levels via microRNA-34a inhibition. J. Biol. Chem. 285:12604–12611; 2010. [254] Lavu, S.; Boss, O.; Elliott, P. J.; Lambert, P. D. Sirtuins—novel therapeutic targets to treat age-associated diseases. Nat. Rev. Drug Discov. 7:841–853; 2008. [255] Villalba, J. M.; de Cabo, R.; Alcain, F. J. A patent review of sirtuin activators: an update. Expert Opin. Ther. Pat. 22:355–367; 2012. [256] Villalba, J. M.; Alcaı´n, F. J. Sirtuin activators and inhibitors. Biofactors 38:349–359; 2012. [257] Jang, M.; Cai, L.; Udeani, G. O.; Slowing, K. V.; Thomas, C. F.; Beecher, C. W.; Fong, H. H.; Farnsworth, N. R.; Kinghorn, A. D.; Mehta, R. G.; Moon, R. C.; Pezzuto, J. M. Cancer chemopreventive activity of resveratrol, a natural product derived from grapes. Science 275:218–220; 1997. [258] Milne, J. C.; Lambert, P. D.; Schenk, S.; Carney, D. P.; Smith, J. J.; Gagne, D. J.; Jin, L.; Boss, O.; Perni, R. B.; Vu, C. B.; Bemis, J. E.; Xie, R.; Disch, J. S.; Ng, P. Y.; Nunes, J. J.; Lynch, A. V.; Yang, H.; Galonek, H.; Israelian, K.; Choy, W.; Iffland, A.; Lavu, S.; Medvedik, O.; Sinclair, D. A.; Olefsky, J. M.; Jirousek, M. R.; Elliott, P. J.; Westphal, C. H. Small molecule activators of SIRT1 as therapeutics for the treatment of type 2 diabetes. Nature 450:712–716; 2007. [259] Minor, R. K.; Baur, J. A.; Gomes, A. P.; Ward, T. M.; Csiszar, A.; Mercken, E. M.; Abdelmohsen, K.; Shin, Y. K.; Canto, C.; Scheibye-Knudsen, M.; Krawczyk, M.; Irusta, P. M.; Martı´n-Montalvo, A.; Hubbard, B. P.; Zhang, Y.; Lehrmann, E.; White, A. A.; Price, N. L.; Swindell, W. R.; Pearson, K. J.; Becker, K. G.; Bohr, V. A.; Gorospe, M.; Egan, J. M.; Talan, M. I.; Auwerx, J.; Westphal, C. H.; Ellis, J. L.; Ungvari, Z.; Vlasuk, G. P.; Elliott, P. J.; Sinclair, D. A.; de Cabo, R. SRT1720 improves survival and healthspan of obese mice. Sci. Rep. 1(article 70):1–10; 2011.

164

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

[260] Beher, D.; Wu, J.; Cumine, S.; Kim, K. W.; Lu, S. C.; Atangan, L.; Wang, M. Resveratrol is not a direct activator of SIRT1 enzyme activity. Chem. Biol. Drug Des. 74:619–624; 2009. [261] Pacholec, M.; Bleasdale, J. E.; Chrunyk, B.; Cunningham, D.; Flynn, D.; Garofalo, R. S.; Griffith, D.; Griffor, M.; Loulakis, P.; Pabst, B.; Qiu, X.; Stockman, B.; Thanabal, V.; Varghese, A.; Ward, J.; Withka, J.; Ahn, K. SRT1720, SRT2183, SRT1460, and resveratrol are not direct activators of SIRT1. J. Biol. Chem. 285:8340–8351; 2010. [262] Kaeberlein, M.; McDonagh, T.; Heltweg, B.; Hixon, J.; Westman, E. A.; Caldwell, S. D.; Napper, A.; Curtis, R.; DiStefano, P. S.; Fields, S.; Bedalov, A.; Kennedy, B. K. Substrate-specific activation of sirtuins by resveratrol. J. Biol. Chem. 280:17038–17045; 2005. [263] Borra, M. T.; Smith, B. C.; Denu, J. M. Mechanism of human SIRT1 activation by resveratrol. J. Biol. Chem. 280:17187–17195; 2005. [264] Dai, H.; Kustigian, L.; Carney, D.; Case, A.; Considine, T.; Hubbard, B. P.; Perni, R. B.; Riera, T. V.; Szczepankiewicz, B.; Vlasuk, G. P.; Stein, R. L. SIRT1 activation by small molecules: kinetic and biophysical evidence for direct interaction of enzyme and activator. J. Biol. Chem. 285:32695–32703; 2010. [265] Baur, J. A.; Pearson, K. J.; Price, N. L.; Jamieson, H. A.; Lerin, C.; Kalra, A.; Prabhu, V. V.; Allard, J. S.; Lopez-Lluch, G.; Lewis, K.; Pistell, P. J.; Poosala, S.; Becker, K. G.; Boss, O.; Gwinn, D.; Wang, M.; Ramaswamy, S.; Fishbein, K. W.; Spencer, R. G.; Lakatta, E. G.; Le Couteur, D. G.; Shaw, R. J.; Navas, P.; Puigserver, P.; Ingram, D. K.; de Cabo, R.; Sinclair, D. A. Resveratrol improves health and survival of mice on a high-calorie diet. Nature 444:337–342; 2006. [266] Zang, M.; Xu, S.; Maitland-Toolan, K. A.; Zuccollo, A.; Hou, X.; Jiang, B.; Wierzbicki, M.; Verbeuren, T. J.; Cohen, R. A. Polyphenols stimulate AMPactivated protein kinase, lower lipids, and inhibit accelerated atherosclerosis in diabetic LDL receptor-deficient mice. Diabetes 55:2180–2191; 2006. [267] Dasgupta, B.; Milbrandt, J. Resveratrol stimulates AMP kinase activity in neurons. Proc. Natl. Acad. Sci. USA 104:7217–7222; 2007. [268] Park, C. E.; Kim, M. J.; Lee, J. H.; Min, B. I.; Bae, H.; Choe, W.; Kim, S. S.; Ha, J. Resveratrol stimulates glucose transport in C2C12 myotubes by activating AMP-activated protein kinase. Exp. Mol. Med. 39:222–229; 2007. [269] Feige, J. N.; Lagouge, M.; Canto, C.; Strehle, A.; Houten, S. M.; Milne, J. C.; Lambert, P. D.; Mataki, C.; Elliott, P. J.; Auwerx, J. Specific SIRT1 activation mimics low energy levels and protects against diet-induced metabolic disorders by enhancing fat oxidation. Cell Metab. 8:347–358; 2008. [270] Um, J. H.; Park, S. J.; Kang, H.; Yang, S.; Foretz, M.; McBurney, M. W.; Kim, M. K.; Viollet, B.; Chung, J. H. AMP-activated protein kinase-deficient mice are resistant to the metabolic effects of resveratrol. Diabetes 59:554–563; 2010. [271] Zheng, J.; Ramirez, V. D. Inhibition of mitochondrial proton F0F1-ATPase/ ATP synthase by polyphenolic phytochemicals. Br. J. Pharmacol. 130:1115–1123; 2000. [272] Gledhill, J. R.; Montgomery, M. G.; Leslie, A. G.; Walker, J. E. Mechanism of inhibition of bovine F1-ATPase by resveratrol and related polyphenols. Proc. Natl. Acad. Sci. USA 104:13632–13637; 2007. [273] Hawley, S. A.; Ross, F. A.; Chevtzoff, C.; Green, K. A.; Evans, A.; Fogarty, S.; Towler, M. C.; Brown, L. J.; Ogunbayo, O. A.; Evans, A. M.; Hardie, D. G. Use of cells expressing gamma subunit variants to identify diverse mechanisms of AMPK activation. Cell Metab. :554–565; 2010. [274] Ruderman, N. B.; Xu, X. J.; Nelson, L.; Cacicedo, J. M.; Saha, A. K.; Lan, F.; Ido, Y. AMPK and SIRT1: a long-standing partnership? Am. J. Physiol. Endocrinol. Metab. 298:E751–E760; 2010. [275] Park, S. -J.; Ahmad, F.; Philp, A.; Baar, K.; Williams, T.; Luo, H.; Ke, H.; Rehmann, H.; Taussig, R.; Brown, A. L.; Kim, M. K.; Beaven, M. A.; Burgin, A. B.; Manganiello, V.; Chung, J. H. Resveratrol ameliorates aging-related metabolic phenotypes by inhibiting cAMP phosphodiesterases. Cell 148:421–433; 2012. [276] Callaway, E. Questions hang over red-wine chemical. How resveratrol benefits health a matter of debate. Nature ; Feb 2, 2012. http://dx.doi.org/ 10.1038/Nature2012.9970. [Epub only]. [277] Canto´, C.; Jiang, L. Q.; Deshmukh, A. S.; Mataki, C.; Coste, A.; Lagouge, M.; Zierath, J. R.; Auwerx, J. Interdependence of AMPK and SIRT1 for metabolic adaptation to fasting and exercise in skeletal muscle. Cell Metab. 11:213–219; 2010. [278] Fulco, M.; Cen, Y.; Zhao, P.; Hoffman, E. P.; McBurney, M. W.; Sauve, A. A.; Sartorelli, V. Glucose restriction inhibits skeletal myoblast differentiation by activating SIRT1 through AMPK-mediated regulation of Nampt. Dev. Cell 14:661–673; 2008. [279] Timmers, S.; Konings, E.; Bilet, L.; Houtkooper, R. H.; van de Weijer, T.; Goossens, G. H.; Hoeks, J.; van der Krieken, S.; Ryu, D.; Kersten, S.; MoonenKornips, E.; Hesselink, M. K.; Kunz, I.; Schrauwen-Hinderling, V. B.; Blaak, E. E.; Auwerx, J.; Schrauwen, P. Calorie restriction-like effects of 30 days of resveratrol supplementation on energy metabolism and metabolic profile in obese humans. Cell Metab. 14:612–622; 2011. [280] Massimi, M.; Tomassini, A.; Sciubba, F.; Sobolev, A. P.; Devirgiliis, L. C.; Miccheli, A. Effects of resveratrol on HepG2 cells as revealed by 1H NMR based metabolic profiling. Biochim. Biophys. Acta 1-8:2012; 1820. [281] Markus, M. A.; Marques, F. Z.; Morris, B. J. Resveratrol, by modulating RNA processing factor levels, can influence the alternative splicing of premRNAs. PLoS One 6(e28926):1–7; 2011. [282] Baur, J. A. Resveratrol sirtuins, and the promise of a DR mimetic. Mech. Ageing Dev. 131:261–269; 2010. [283] Pearson, K. J.; Baur, J. A.; Lewis, K. N.; Peshkin, L.; Price, N. L.; Labinskyy, N.; Swindell, W. R.; Kamara, D.; Minor, R. K.; Perez, E.; Jamieson, H. A.; Zhang,

[284]

[285]

[286]

[287]

[288]

[289]

[290]

[291]

[292]

[293]

[294]

[295]

[296]

[297]

[298]

[299] [300]

[301]

[302]

[303]

[304]

[305]

[306]

Y.; Dunn, S. R.; Sharma, K.; Pleshko, N.; Woollett, L. A.; Csiszar, A.; Ikeno, Y.; Le Couteur, D.; Elliott, P. J.; Becker, K. G.; Navas, P.; Ingram, D. K.; Wolf, N. S.; Ungvari, Z.; Sinclair, D. A.; de Cabo, R. Resveratrol delays age-related deterioration and mimics transcriptional aspects of dietary restriction without extending life span. Cell Metab. 8:157–168; 2008. Yamazaki, Y.; Usui, I.; Kanatani, Y.; Matsuya, Y.; Tsuneyama, K.; Fujisaka, S.; Bukhari, A.; Suzuki, H.; Senda, S.; Imanishi, S.; Hirata, K.; Ishiki, M.; Hayashi, R.; Urakaze, M.; Nemoto, H.; Kobayashi, M.; Tobe, K. Treatment with SRT1720, a SIRT1 activator, ameliorates fatty liver with reduced expression of lipogenic enzymes in MSG mice. Am. J. Physiol. Endocrinol. Metab. 297:E1179–E1186; 2009. Huber, J. L.; McBurney, M. W.; Distefano, P. S.; McDonagh, T. SIRT1independent mechanisms of the putative sirtuin enzyme activators SRT1720 and SRT2183. Future Med. Chem 2:1751–1759; 2010. Revollo, J. R.; Grimm, A. A.; Imai, S. The NAD biosynthesis pathway mediated by nicotinamide phosphoribosyltransferase regulates Sir2 activity in mammalian cells. J. Biol. Chem. 279:50754–50763; 2004. Vaccari, C. S.; Nagamia, S.; Thoenes, M.; Oguchi, A.; Hammoud, R.; Khan, B. V. Efficacy of controlled-release niacin in treatment of metabolic syndrome: correlation to surrogate markers of atherosclerosis, vascular reactivity, and inflammation. J. Clin. Lipidol 1:605–613; 2007. Westphal, S.; Borucki, K.; Taneva, E.; Makarova, R.; Luley, C. Extendedrelease niacin raises adiponectin and leptin. Atherosclerosis 193:361–365; 2007. Huang, Y. N.; Qi, J. H.; Xiang, L.; Wang, Y. Z. Construction of adiponectinencoding plasmid DNA and overexpression in mice in vivo. Gene 502:87–93; 2012. Canto, C.; Houtkooper, R. H.; Pirinen, E.; Youn, D. Y.; Oosterveer, M. H.; Cen, Y.; Fernandez-Marcos, P. J.; Yamamoto, H.; Andreux, P. A.; Cettour-Rose, P.; Gademann, K.; Rinsch, C.; Schoonjans, K.; Sauve, A. A.; Auwerx, J. The NAD þ precursor nicotinamide riboside enhances oxidative metabolism and protects against high-fat diet-induced obesity. Cell Metab. 15:838–847; 2012. van der Veer, E.; Nong, Z.; O’Neil, C.; Urquhart, B.; Freeman, D.; Pickering, J. G. Pre-B-cell colony-enhancing factor regulates NAD þ -dependent protein deacetylase activity and promotes vascular smooth muscle cell maturation. Circ. Res. 97:25–34; 2005. van der Veer, E.; Ho, C.; O’Neil, C.; Barbosa, N.; Scott, R.; Cregan, S. P.; Pickering, J. G. Extension of human cell lifespan by nicotinamide phosphoribosyltransferase. J. Biol. Chem. 282:10841–10845; 2007. ¨ Revollo, J. R.; Korner, A.; Mills, K. F.; Satoh, A.; Wang, T.; Garten, A.; Dasgupta, B.; Sasaki, Y.; Wolberger, C.; Townsend, R. R.; Milbrandt, J.; Kiess, W.; Imai, S. Nampt/PBEF/Visfatin regulates insulin secretion in beta cells as a systemic NAD biosynthetic enzyme. Cell Metab. 6:363–375; 2007. Pittelli, M.; Formentini, L.; Faraco, G.; Lapucci, A.; Rapizzi, E.; Cialdai, F.; Romano, G.; Moneti, G.; Moroni, F.; Chiarugi, A. Inhibition of nicotinamide phosphoribosyltransferase: cellular bioenergetics reveals a mitochondrial insensitive NAD pool. J. Biol. Chem. 285:34106–34114; 2010. Pillai, J. B.; Isbatan, A.; Imai, S.; Gupta, M. P. Poly(ADP-ribose) polymerase-1dependent cardiac myocyte cell death during heart failure is mediated by NAD þ depletion and reduced Sir2a deacetylase activity. J. Biol. Chem. 280:43121–43130; 2005. Mendoza-Alvarez, H.; Alvarez-Gonzalez, R. Poly(ADP-ribose) polymerase is a catalytic dimer and the automodification reaction is intermolecular. J. Biol. Chem 268:22575–22580; 1993. Yuan, Y.; Liao, Y. M.; Hsueh, C. T.; Mirshahidi, H. R. Novel targeted therapeutics: inhibitors of MDM2, ALK and PARP. J. Hematol. Oncol. 4(article 16):1–14; 2011. Lim, J. H.; Lee, Y. M.; Chun, Y. S.; Chen, J.; Kim, J. E.; Park, J. W. Sirtuin 1 modulates cellular responses to hypoxia by deacetylating hypoxiainducible factor 1(a). Mol. Cell 38:864–878; 2010. Canto, C.; Auwerx, J. Interference between PARPs and SIRT1: a novel approach to healthy ageing? Aging (Albany NY) 3:543–547; 2011. Mangerich, A.; Herbach, N.; Hanf, B.; Fischbach, A.; Popp, O.; Moreno¨ Villanueva, M.; Bruns, O. T.; Burkle, A. Inflammatory and age-related pathologies in mice with ectopic expression of human PARP-1. Mech. Ageing Dev. 131:389–404; 2010. Yang, H.; Yang, T.; Baur, J. A.; Perez, E.; Matsui, T.; Carmona, J. J.; Lamming, D. W.; Souza-Pinto, N. C.; Bohr, V. A.; Rosenzweig, A.; de Cabo, R.; Sauve, A. A.; Sinclair, D. A. Nutrient-sensitive mitochondrial NAD þ levels dictate cell survival. Cell 130:1095–1107; 2007. Cakir-Kiefer, C.; Muller-Steffner, H.; Oppenheimer, N.; Schuber, F. Kinetic competence of the cADP-ribose-CD38 complex as an intermediate in the CD38/NAD þ glycohydrolase-catalysed reactions: implication for CD38 signalling. Biochem. J 358:399–406; 2001. Dousa, T. P.; Chini, E. N.; Beers, K. W. Adenine nucleotide diphosphates: emerging second messengers acting via intracellular Ca2 þ release. Am. J. Physiol. Endocrinol. Metab. 271:C1007–C1024; 1996. Yamada, M.; Mizuguchi, M.; Otsuka, N.; Ikeda, K.; Takahashi, H. Ultrastructural localization of CD38 immunoreactivity in rat brain. Brain Res. 756:52–60; 1997. Aksoy, P.; White, T. A.; Thompson, M.; Chini, E. N. Regulation of intracellular levels of NAD: a novel role for CD38. Biochem. Biophys. Res. Commun. 345:1386–1392; 2006. Aksoy, P.; Escande, C.; White, T. A.; Thompson, M.; Soares, S.; Benech, J. C.; Chini, E. N. Regulation of SIRT 1 mediated NAD dependent deacetylation: a

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

[307]

[308]

[309]

[310]

[311] [312]

[313]

[314]

[315]

[316]

[317]

[318]

[319]

[320]

[321]

[322]

[323] [324]

[325] [326]

[327]

[328]

[329]

novel role for the multifunctional enzyme CD38. Biochem. Biophys. Res. Commun. 349:353–359; 2006. Dong, M.; Si, Y. Q.; Sun, S. Y.; Pu, X. P.; Yang, Z. J.; Zhang, L. R.; Zhang, L. H.; Leung, F. P.; Lam, C. M.; Kwong, A. K.; Yue, J.; Zhou, Y.; Kriksunov, I. A.; Hao, Q.; Lee, H. C. Design, synthesis and biological characterization of novel inhibitors of CD38. Org. Biomol. Chem. 9:3246–3257; 2011. Barger, J. L.; Kayo, T.; Vann, J. M.; Arias, E. B.; Wang, J.; Hacker, T. A.; Wang, Y.; Raederstorff, D.; Morrow, J. D.; Leeuwenburgh, C.; Allison, D. B.; Saupe, K. W.; Cartee, G. D.; Weindruch, R.; Prolla, T. A. A low dose of dietary resveratrol partially mimics caloric restriction and retards aging parameters in mice. PLoS One 3:e2264; 2008. Smith, J. J.; Kenney, R. D.; Gagne, D. J.; Frushour, B. P.; Ladd, W.; Galonek, H. L.; Israelian, K.; Song, J.; Razvadauskaite, G.; Lynch, A. V.; Carney, D. P.; Johnson, R. J.; Lavu, S.; Iffland, A.; Elliott, P. J.; Lambert, P. D.; Elliston, K. O.; Jirousek, M. R.; Milne, J. C.; Boss, O. Small molecule activators of SIRT1 replicate signaling pathways triggered by calorie restriction in vivo. BMC Syst. Biol. 3:31; 2009. Price, N. L.; Gomes, A. P.; Ling, A. J.; Duarte, F. V.; Martin-Montalvo, A.; North, B. J.; Agarwal, B.; Ye, L.; Ramadori, G.; Teodoro, J. S.; Hubbard, B. P.; Varela, A. T.; Davis, J. G.; Varamini, B.; Hafner, A.; Moaddel, R.; Rolo, A. P.; Coppari, R.; Palmeira, C. M.; de Cabo, R.; Baur, J. A.; Sinclair, D. A. SIRT1 is required for AMPK activation and the beneficial effects of resveratrol on mitochondrial function. Cell Metab. 15:675–690; 2012. Denu, J. M. Fortifying the link between SIRT1, resveratrol, and mitochondrial function. Cell Metab. 15:566–567; 2012. Centeno-Baez, C.; Dallaire, P.; Marette, A. Resveratrol inhibition of inducible nitric oxide synthase in skeletal muscle involves AMPK but not SIRT1. Am. J. Physiol. Endocrinol. Metab. 301:E922–E930; 2011. Bemis, J. E.; Vu, C. B.; Xie, R.; Nunes, J. J.; Ng, P. Y.; Disch, J. S.; Milne, J. C.; Carney, D. P.; Lynch, A. V.; Jin, L.; Smith, J. J.; Lavu, S.; Iffland, A.; Jirousek, M. R.; Perni, R. B. Discovery of oxazolo[4,5-b]pyridines and related heterocyclic analogs as novel SIRT1 activators. Bioorg. Med. Chem. Lett. 19:2350–2353; 2009. Vu, C. B.; Bemis, J. E.; Disch, J. S.; Ng, P. Y.; Nunes, J. J.; Milne, J. C.; Carney, D. P.; Lynch, A. V.; Smith, J. J.; Lavu, S.; Lambert, P. D.; Gagne, D. J.; Jirousek, M. R.; Schenk, S.; Olefsky, J. M.; Perni, R. B. Discovery of imidazo[1,2-b]thiazole derivatives as novel SIRT1 activators. J. Med. Chem. 52:1275–1283; 2009. Fride´n-Saxin, M.; Seifert, T.; Landergren, M. R.; Suuronen, T.; LahtelaKakkonen, M.; Jarho, E. M.; Luthman, K. Synthesis and evaluation of substituted chroman-4-one and chromone derivatives as sirtuin 2selective inhibitors. J. Med. Chem. ; August 7, 2012. [Epub ahead of print]. Galli, U.; Mesenzani, O.; Coppo, C.; Sorba, G.; Canonico, P. L.; Tron, G. C.; Genazzani, A. A. Identification of a sirtuin 3 inhibitor that displays selectivity over sirtuin 1 and 2. Eur. J. Med. Chem. 55:58–66; 2012. Mai, A.; Valente, S.; Meade, S.; Carafa, V.; Tardugno, M.; Nebbioso, A.; Galmozzi, A.; Mitro, N.; De Fabiani, E.; Altucci, L.; Kazantsev, A. Study of 1,4dihydropyridine structural scaffold: discovery of novel sirtuin activators and inhibitors. J. Med. Chem. 52:5496–5504; 2009. Fu, J.; Jin, J.; Cichewicz, R. H.; Hageman, S. A.; Ellis, T. K.; Xiang, L.; Peng, Q.; Jiang, M.; Arbez, N.; Hotaling, K.; Ross, C. A.; Duan, W. Trans-(-)-e-viniferin increases mitochondrial sirtuin 3 (SIRT3), activates AMPK, and protects cells in models of Huntington’s disease. J. Biol. Chem. 287:24460–24472; 2012. Smoliga, J. M.; Vang, O.; Baur, J. A. Challenges of translating basic research into therapeutics: resveratrol as an example. J. Gerontol. A Biol. Sci. Med. Sci 67:158–167; 2012. Dietrich, M. O.; Antunes, C.; Geliang, G.; Liu, Z. W.; Borok, E.; Nie, Y.; Xu, A. W.; Souza, D. O.; Gao, Q.; Diano, S.; Gao, X. B.; Horvath, T. L. Agrp neurons mediate Sirt1’s action on the melanocortin system and energy balance: roles for Sirt1 in neuronal firing and synaptic plasticity. J. Neurosci. 30:11815–11825; 2010. Cakir, I.; Perello, M.; Lansari, O.; Messier, N. J.; Vaslet, C. A.; Nillni, E. A. Hypothalamic Sirt1 regulates food intake in a rodent model system. PLoS One 4(article e8322):1–12; 2009. Ramadori, G.; Fujikawa, T.; Fukuda, M.; Anderson, J.; Morgan, D. A.; Mostoslavsky, R.; Stuart, R. C.; Perello, M.; Vianna, C. R.; Nillni, E. A.; Rahmouni, K.; Coppari, R. SIRT1 deacetylase in POMC neurons is required for homeostatic defenses against diet-induced obesity. Cell Metab. 12:78–87; 2010. Gillum, M. P.; Erion, D. M.; Shulman, G. I. Sirtuin-1 regulation of mammalian metabolism. Trends Mol. Med. 17:8–13; 2010. Ramadori, G.; Fujikawa, T.; Anderson, J.; Berglund, E. D.; Frazao, R.; Micha´n, S.; Vianna, C. R.; Sinclair, D. A.; Elias, C. F.; Coppari, R. SIRT1 deacetylase in SF1 neurons protects against metabolic imbalance. Cell Metab. 14:301–312; 2011. Vianna, C. R.; Coppari, R. A treasure trove of hypothalamic neurocircuitries governing body weight homeostasis. Endocrinology 152:11–18; 2011. Wu, D.; Qiu, Y.; Gao, X.; Yuan, X. B.; Zhai, Q. Overexpression of SIRT1 in mouse forebrain impairs lipid/glucose metabolism and motor function. PLoS One 6:e21759; 2011. Xu, F.; Burk, D.; Gao, Z.; Yin, J.; Zhang, X.; Weng, J.; Ye, J. Angiogenic deficiency and adipose tissue dysfunction are associated with macrophage malfunction in SIRT1  /  mice. Endocrinology 153:1706–1716; 2012. Gillum, M. P.; Kotas, M. E.; Erion, D. M.; Kursawe, R.; Chatterjee, P.; Nead, K. T.; Muise, E. S.; Hsiao, J. J.; Frederick, D. W.; Yonemitsu, S.; Banks, A. S.; Qiang, L.; Bhanot, S.; Olefsky, J. M.; Sears, D. D.; Caprio, S.; Shulman, G. I. SirT1 regulates adipose tissue inflammation. Diabetes 60:3235–3245; 2011. Xu, F.; Gao, Z.; Zhang, J.; Rivera, C. A.; Yin, J.; Weng, J.; Ye, J. Lack of SIRT1 (Mammalian Sirtuin 1) activity leads to liver steatosis in the SIRT1 þ / mice: a

[330]

[331]

[332]

[333]

[334]

[335]

[336]

[337]

[338]

[339]

[340]

[341]

[342]

[343]

[344]

[345]

[346]

[347]

[348]

[349]

[350]

165

role of lipid mobilization and inflammation. Endocrinology 151:2504–2514; 2010. Lee, J.; Hong, S. W.; Chae, S. W.; Kim, D. H.; Choi, J. H.; Bae, J. C.; Park, S. E.; Rhee, E. J.; Park, C. Y.; Oh, K. W.; Park, S. W.; Kim, S. W.; Lee, W. Y. Exendin-4 improves steatohepatitis by increasing Sirt1 expression in high-fat dietinduced obese C57BL/6J mice. PLoS One 7:e31394; 2012. Chakrabarti, P.; English, T.; Karki, S.; Qiang, L.; Tao, R.; Kim, J.; Luo, Z.; Farmer, S. R.; Kandror, K. V. SIRT1 controls lipolysis in adipocytes via FOXO1-mediated expression of ATGL. J. Lipid Res. 52:1693–1701; 2011. Krishnan, J.; Danzer, C.; Simka, T.; Ukropec, J.; Walter, K. M.; Kumpf, S.; Mirtschink, P.; Ukropcova, B.; Gasperikova, D.; Pedrazzini, T.; Krek, W. Dietary obesity-associated Hif1a activation in adipocytes restricts fatty acid oxidation and energy expenditure via suppression of the Sirt2-NAD þ system. Genes Dev. 26:259–270; 2012. Chen, W. L.; Kang, C. H.; Wang, S. G.; Lee, H. M. a-Lipoic acid regulates lipid metabolism through induction of sirtuin 1 (SIRT1) and activation of AMPactivated protein kinase. Diabetologia 55:824–835; 2012. Valdecantos, M. P.; Pe´rez-Matute, P.; Gonza´lez-Muniesa, P.; Prieto-Hontoria, P. L.; Moreno-Aliaga, M. J.; Martı´nez, J. A. Lipoic acid improves mitochondrial function in nonalcoholic steatosis through the stimulation of sirtuin 1 and sirtuin 3. Obesity (Silver Spring) ; February 13, 2012. [Epub ahead of print]. Strum, J. C.; Johnson, J. H.; Ward, J.; Xie, H.; Feild, J.; Hester, A.; Alford, A.; Waters, K. M. MicroRNA 132 regulates nutritional stress-induced chemokine production through repression of SirT1. Mol. Endocrinol. 23: 1876–1884; 2009. Castro, R. E.; Ferreira, D. M.; Afonso, M. B.; Borralho, P. M.; Machado, M. V.; Cortez-Pinto, H.; Rodrigues, C. M. miR-34a/SIRT1/p53 is suppressed by ursodeoxycholic acid in rat liver and activated by disease severity in human nonalcoholic fatty liver disease. J. Hepatol. ; August 15, 2012. [Epub ahead of print]. Borengasser, S.; Lau, F.; Kang, P.; Blackburn, M. L.; Ronis, M. J.; Badger, T. M.; Shankar, K. Maternal obesity during gestation impairs fatty acid oxidation and mitochondrial SIRT3 expression in rat offspring at weaning. PLoS One 6:e24068; 2011. Knight, C. M.; Gutierrez-Juarez, R.; Lam, T. K.; Arrieta-Cruz, I.; Huang, L.; Schwartz, G.; Barzilai, N.; Rossetti, L. Mediobasal hypothalamic SIRT1 is essential for resveratrol’s effects on insulin action in rats. Diabetes 60:2691–2700; 2011. Sun, C.; Zhang, F.; Ge, X.; Yan, T.; Chen, X.; Shi, X.; Zhai, Q. SIRT1 improves insulin sensitivity under insulin-resistant conditions by repressing PTP1B. Cell Metab. 6:307–319; 2007. Nelson, L. E.; Valentine, R. J.; Cacicedo, J. M.; Gauthier, M. S.; Ido, Y.; Ruderman, N. B. A novel inverse relationship between metformin-triggered AMPK-SIRT1 signaling and p53 protein abundance in high glucose exposed HepG2 cells. Am. J. Physiol. Cell Physiol 303:C4–C13; 2012. Zheng, Z.; Chen, H.; Li, J.; Li, T.; Zheng, B.; Zheng, Y.; Jin, H.; He, Y.; Gu, Q.; Xu, X. Sirtuin 1-mediated cellular metabolic memory of high glucose via the LKB1/AMPK/ROS pathway and therapeutic effects of metformin. Diabetes 61:217–228; 2012. Kubota, S.; Ozawa, Y.; Kurihara, T.; Sasaki, M.; Yuki, K.; Miyake, S.; Noda, K.; Ishida, S.; Tsubota, K. Roles of AMP-activated protein kinase in diabetesinduced retinal inflammation. Invest. Ophthalmol. Vis. Sci. 52:9142–9148; 2011. Uribarri, J.; Cai, W.; Ramdas, M.; Goodman, S.; Pyzik, R.; Chen, X.; Zhu, L.; Striker, G. E.; Vlassara, H. Restriction of advanced glycation end products improves insulin resistance in human type 2 diabetes: potential role of AGER1 and SIRT1. Diabetes Care 34:1610–1616; 2011. Li, N.; Muthusamy, S.; Liang, R.; Sarojini, H.; Wang, E. Increased expression of miR-34a and miR-93 in rat liver during aging, and their impact on the expression of Mgst1 and Sirt1. Mech. Ageing Dev 132:75–85; 2011. Zhou, B.; Li, C.; Qi, W.; Zhang, Y.; Zhang, F.; Wu, J. X.; Hu, Y. N.; Wu, D. M.; Liu, Y.; Yan, T. T.; Jing, Q.; Liu, M. F.; Zhai, Q. W. Downregulation of miR181a upregulates sirtuin-1 (SIRT1) and improves hepatic insulin sensitivity. Diabetologia 55:2032–2043; 2012. Granata, R.; Gallo, D.; Luque, R. M.; Baragli, A.; Scarlatti, F.; Grande, C.; Gesmundo, I.; Co´rdoba-Chaco´n, J.; Bergandi, L.; Settanni, F.; Togliatto, G.; Volante, M.; Garetto, S.; Annunziata, M.; Chanclo´n, B.; Gargantini, E.; Rocchietto, S.; Matera, L.; Datta, G.; Morino, M.; Brizzi, M. F.; Ong, H.; Camussi, G.; Castan˜o, J. P.; Papotti, M.; Ghigo, E. Obestatin regulates adipocyte function and protects against diet-induced insulin resistance and inflammation. FASEB J 26:3393–3411; 2012. Kitada, M.; Takeda, A.; Nagai, T.; Ito, H.; Kanasaki, K.; Koya, D. Dietary restriction ameliorates diabetic nephropathy through anti-inflammatory effects and regulation of the autophagy via restoration of Sirt1 in diabetic Wistar fatty (fa/fa) rats: a model of type 2 diabetes. Exp. Diabetes Res 2011:908185; 2011. Xu, Y.; Nie, L.; Yin, Y. G.; Tang, J. L.; Zhou, J. Y.; Li, D. D.; Zhou, S. W. Resveratrol protects against hyperglycemia-induced oxidative damage to mitochondria by activating SIRT1 in rat mesangial cells. Toxicol. Appl. Pharmacol. 259:395–401; 2012. Zhang, S.; Cai, G.; Fu, B.; Liu, W.; Zhuo, L.; Sun, L.; Liu, F.; Chen, X. SIRT1 is required for the effects of rapamycin on high glucose-inducing mesangial cells senescence. Mech. Ageing Dev. 133:387–400; 2012. Chuang, P. Y.; Dai, Y.; Liu, R.; He, H.; Kretzler, M.; Jim, B.; Cohen, C. D.; He, J. C. Alteration of forkhead box O (foxo4) acetylation mediates apoptosis of podocytes in diabetes mellitus. PLoS One 6:e23566; 2011.

166

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

[351] Yuan, Y.; Huang, S.; Wang, W.; Wang, Y.; Zhang, P.; Zhu, C.; Ding, G.; Liu, B.; Yang, T.; Zhang, A. Activation of peroxisome proliferator-activated receptorg coactivator 1a ameliorates mitochondrial dysfunction and protects podocytes from aldosterone-induced injury. Kidney Int. 82:771–789; 2012. [352] Dolinsky, V. W.; Dyck, J. R. Calorie restriction and resveratrol in cardiovascular health and disease. Biochim. Biophys. Acta 1477-1489:2011; 1812. [353] Oellerich, M. F.; Potente, M. FOXOs and sirtuins in vascular growth, maintenance, and aging. Circ. Res. 110:1238–1251; 2012. [354] Fontana, L.; Meyer, T. E.; Klein, S.; Holloszy, J. O. Long-term calorie restriction is highly effective in reducing the risk for atherosclerosis in humans. Proc. Natl. Acad. Sci. USA 101:6659–6663; 2004. [355] Nisoli, E.; Tonello, C.; Cardile, A.; Cozzi, V.; Bracale, R.; Tedesco, L.; Falcone, S.; Valerio, A.; Cantoni, O.; Clementi, E.; Moncada, S.; Carruba, M. O. Calorie restriction promotes mitochondrial biogenesis by inducing the expression of eNOS. Science 310:314–317; 2005. [356] Stein, S.; Matter, C. M. Protective roles of SIRT1 in atherosclerosis. Cell Cycle 10:640–647; 2011. [357] Zhang, Q. J.; Wang, Z.; Chen, H. Z.; Zhou, S.; Zheng, W.; Liu, G.; Wei, Y. S.; Cai, H.; Liu, D. P.; Liang, C. C. Endothelium-specific overexpression of class III deacetylase SIRT1 decreases atherosclerosis in apolipoprotein E-deficient mice. Cardiovasc. Res. 80:191–199; 2008. [358] Do, G. M.; Kwon, E. Y.; Kim, H. J.; Jeon, S. M.; Ha, T. Y.; Park, T.; Choi, M. S. Long-term effects of resveratrol supplementation on suppression of atherogenic lesion formation and cholesterol synthesis in apo E-deficient mice. Biochem. Biophys. Res. Commun. 374:55–59; 2008. [359] Chen, Z.; Peng, I. C.; Cui, X.; Li, Y. S.; Chien, S.; Shyy, J. Y. Shear stress, SIRT1, and vascular homeostasis. Proc. Natl. Acad. Sci. USA 107:10268–10273; 2010. [360] Tajbakhsh, N.; Sokoya, E. M. Regulation of cerebral vascular function by sirtuin 1. Microcirculation 19:336–342; 2012. ¨ [361] Stein, S.; Lohmann, C.; Schafer, N.; Hofmann, J.; Rohrer, L.; Besler, C.; Rothgiesser, K. M.; Becher, B.; Hottiger, M. O.; Bore´n, J.; McBurney, M. W.; ¨ Landmesser, U.; Luscher, T. F.; Matter, C. M. SIRT1 decreases Lox-1mediated foam cell formation in atherogenesis. Eur. Heart J. 31:2301–2309; 2011. [362] Hasegawa, Y.; Saito, T.; Ogihara, T.; Ishigaki, Y.; Yamada, T.; Imai, J.; Uno, K.; Gao, J.; Kaneko, K.; Shimosawa, T.; Asano, T.; Fujita, T.; Oka, Y.; Katagiri, H. Blockade of the nuclear factor-kB pathway in the endothelium prevents insulin resistance and prolongs life spans. Circulation 125:1122–1133; 2012. [363] Potente, M.; Ghaeni, L.; Baldessari, D.; Mostoslavsky, R.; Rossig, L.; Dequiedt, F.; Haendeler, J.; Mione, M.; Dejana, E.; Alt, F. W.; Zeiher, A. M.; Dimmeler, S. SIRT1 controls endothelial angiogenic functions during vascular growth. Genes Dev. 21:2644–2658; 2007. [364] Paik, J. H.; Kollipara, R.; Chu, G.; Ji, H.; Xiao, Y.; Ding, Z.; Miao, L.; Tothova, Z.; Horner, J. W.; Carrasco, D. R.; Jiang, S.; Gilliland, D. G.; Chin, L.; Wong, W. H.; Castrillon, D. H.; DePinho, R. A. FoxOs are lineage-restricted redundant tumor suppressors and regulate endothelial cell homeostasis. Cell 128:309–323; 2007. ¨ [365] Guarani, V.; Deflorian, G.; Franco, C. A.; Kruger, M.; Phng, L. K.; Bentley, K.; Toussaint, L.; Dequiedt, F.; Mostoslavsky, R.; Schmidt, M. H.; Zimmermann, B.; Brandes, R. P.; Mione, M.; Westphal, C. H.; Braun, T.; Zeiher, A. M.; Gerhardt, H.; Dimmeler, S.; Potente, M. Acetylation-dependent regulation of endothelial Notch signalling by the SIRT1 deacetylase. Nature 473:234–238; 2011. [366] Sciacca, S.; Pilato, M.; Mazzoccoli, G.; Pazienza, V.; Vinciguerra, M. Anticorrelation between longevity gene SirT1 and Notch signaling in ascending aorta biopsies from patients with bicuspid aortic valve disease. Heart Vessels; February 28, 2012. [Epub ahead of print]. [367] Hashimoto-Komatsu, A.; Hirase, T.; Asaka, M.; Node, K. Angiotensin II induces microtubule reorganization mediated by a deacetylase SIRT2 in endothelial cells. Hypertens. Res. 34:949–956; 2011. ˜ a, G. Activation of [368] Gracia-Sancho, J.; Villarreal Jr G.; Zhang, Y.; Garcı´a-Carden SIRT1 by resveratrol induces KLF2 expression conferring an endothelial vasoprotective phenotype. Cardiovasc. Res. 85:514–519; 2010. [369] Xia, L.; Ding, F.; Zhu, J. H.; Fu, G. S. Resveratrol attenuates apoptosis of pulmonary microvascular endothelial cells induced by high shear stress and proinflammatory factors. Hum. Cell 24:127–133; 2011. [370] Miyazaki, R.; Ichiki, T.; Hashimoto, T.; Inanaga, K.; Imayama, I.; Sadoshima, J.; Sunagawa, K. SIRT1, a longevity gene, downregulates angiotensin II type 1 receptor expression in vascular smooth muscle cells. Arterioscler. Thromb. Vasc. Biol. 28:1263–1269; 2008. [371] Benigni, A.; Corna, D.; Zoja, C.; Sonzogni, A.; Latini, R.; Salio, M.; Conti, S.; Rottoli, D.; Longaretti, L.; Cassis, P.; Morigi, M.; Coffman, T. M.; Remuzzi, G. Disruption of the Ang II type 1 receptor promotes longevity in mice. J. Clin. Invest. 119:524–530; 2009. [372] Chen, R.; Dioum, E. M.; Hogg, R. T.; Gerard, R. D.; Garcia, J. A. Hypoxia increases sirtuin 1 expression in a hypoxia-inducible factor-dependent manner. J. Biol. Chem. 286:13869–13878; 2011. [373] Dioum, E. M.; Chen, R.; Alexander, M. S.; Zhang, Q.; Hogg, R. T.; Gerard, R. D.; Garcia, J. A. Regulation of hypoxia-inducible factor 2a signaling by the stress-responsive deacetylase sirtuin 1. Science 324:1289–1293; 2009. [374] Majmundar, A. J.; Wong, W. J.; Simon, M. C. Hypoxia-inducible factors and the response to hypoxic stress. Mol. Cell 40:294–309; 2010. [375] Balaiya, S.; Khetpal, V.; Chalam, K. V. Hypoxia initiates sirtuin1-mediated vascular endothelial growth factor activation in choroidal endothelial cells through hypoxia inducible factor-2a. Mol. Vis 18:114–120; 2012.

[376] Ota, H.; Eto, M.; Kano, M. R.; Ogawa, S.; Iijima, K.; Akishita, M.; Ouchi, Y. Cilostazol inhibits oxidative stress-induced premature senescence via upregulation of Sirt1 in human endothelial cells. Arterioscler. Thromb. Vasc. Biol. 28:1634–1639; 2008. [377] Zu, Y.; Liu, L.; Lee, M. Y.; Xu, C.; Liang, Y.; Man, R. Y.; Vanhoutte, P. M.; Wang, Y. SIRT1 promotes proliferation and prevents senescence through targeting LKB1 in primary porcine aortic endothelial cells. Circ. Res. 106:1384–1393; 2010. [378] Zhou, S.; Chen, H. Z.; Wan, Y. Z.; Zhang, Q. J.; Wei, Y. S.; Huang, S.; Liu, J. J.; Lu, Y. B.; Zhang, Z. Q.; Yang, R. F.; Zhang, R.; Cai, H.; Liu, D. P.; Liang, C. C. Repression of P66Shc expression by SIRT1 contributes to the prevention of hyperglycemia-induced endothelial dysfunction. Circ. Res. 109:639–648; 2011. [379] Olmos, Y.; Valle, I.; Borniquel, S.; Tierrez, A.; Soria, E.; Lamas, S.; Monsalve, M. Mutual dependence of Foxo3a and PGC-1a in the induction of oxidative stress genes. J. Biol. Chem. 284:14476–14484; 2009. [380] Wang, P.; Guan, Y. F.; Du, H.; Zhai, Q. W.; Su, D. F.; Miao, C. Y. Induction of autophagy contributes to the neuroprotection of nicotinamide phosphoribosyltransferase in cerebral ischemia. Autophagy 8:77–87; 2012. [381] Lemarie´, C. A.; Shbat, L.; Marchesi, C.; Angulo, O. J.; Deschˆenes, M. E.; Blostein, M. D.; Paradis, P.; Schiffrin, E. L. Mthfr deficiency induces endothelial progenitor cell senescence via uncoupling of eNOS and downregulation of SIRT1. Am. J. Physiol. Heart Circ. Physiol 300:H745–H753; 2011. [382] Ito, T.; Yagi, S.; Yamakuchi, M. MicroRNA-34a regulation of endothelial senescence. Biochem. Biophys. Res. Commun. 398:735–740; 2010. [383] Zhao, T.; Li, J.; Chen, A. F. MicroRNA-34a induces endothelial progenitor cell senescence and impedes its angiogenesis via suppressing silent information regulator 1. Am. J. Physiol. Endocrinol. Metab. 299:E110–E116; 2010. [384] Tabuchi, T.; Satoh, M.; Itoh, T.; Nakamura, M. MicroRNA-34a regulates the longevity-associated protein SIRT1 in coronary artery disease: effect of statins on SIRT1 and microRNA-34a expression. Clin. Sci. (Lond.) 123:161–171; 2012. [385] Chen, J.; Xavier, S.; Moskowitz-Kassai, E.; Chen, R.; Lu, C. Y.; Sanduski, K.; ˇ Spes, A.; Turk, B.; Goligorsky, M. S. Cathepsin cleavage of sirtuin 1 in endothelial progenitor cells mediates stress-induced premature senescence. Am. J. Pathol. 180:973–983; 2012. [386] Li, X.; Zhang, S.; Blander, G.; Tse, J. G.; Krieger, M.; Guarente, L. SIRT1 deacetylates and positively regulates the nuclear receptor LXR. Mol. Cell 28:91–106; 2007. [387] Kemper, J. K.; Xiao, Z.; Ponugoti, B.; Miao, J.; Fang, S.; Kanamaluru, D.; Tsang, S.; Wu, S. Y.; Chiang, C. M.; Veenstra, T. D. FXR acetylation is normally dynamically regulated by p300 and SIRT1 but constitutively elevated in metabolic disease states. Cell Metab. 10:392–404; 2009. [388] Qiang, L.; Lin, H. V.; Kim-Muller, J. Y.; Welch, C. L.; Gu, W.; Accili, D. Proatherogenic abnormalities of lipid metabolism in SirT1 transgenic mice are mediated through Creb deacetylation. Cell Metab. 14:758–767; 2011. [389] Planavila, A.; Iglesias, R.; Giralt, M.; Villarroya, F. Sirt1 acts in association with PPARa to protect the heart from hypertrophy, metabolic dysregulation, and inflammation. Cardiovasc. Res. 90:276–284; 2011. [390] Oka, S.; Alcendor, R.; Zhai, P.; Park, J. Y.; Shao, D.; Cho, J.; Yamamoto, T.; Tian, B.; Sadoshima, J. PPARa-Sirt1 complex mediates cardiac hypertrophy and failure through suppression of the ERR transcriptional pathway. Cell Metab. 14:598–611; 2011. [391] Jian, B. Y. S.; Chaudry, I. H.; Raju, R. Resveratrol improves cardiac contractility following trauma-hemorrhage by modulating Sirt1. Mol. Med. 18:209–214; 2012. [392] Nadtochiy, S. M.; Yao, H.; McBurney, M. W.; Gu, W.; Guarente, L.; Rahman, I.; Brookes, P. S. SIRT1-mediated acute cardioprotection. Am. J. Physiol. Heart Circ. Physiol 301:H1506–H1512; 2011. [393] Vinciguerra, M.; Santini, M. P.; Martinez, C.; Pazienza, V.; Claycomb, W. C.; Giuliani, A.; Rosenthal, N. mIGF-1/JNK1/SirT1 signaling confers protection against oxidative stress in the heart. Aging Cell 11:139–149; 2012. [394] Parodi-Rullan, R.; Barreto-Torres, G.; Ruiz, L.; Casasnovas, J.; Javadov, S. Direct renin inhibition exerts an anti-hypertrophic effect associated with improved mitochondrial function in post-infarction heart failure in diabetic rats. Cell Physiol. Biochem 29:841–850; 2012. [395] Sack, M. N. The role of SIRT3 in mitochondrial homeostasis and cardiac adaptation to hypertrophy and aging. J. Mol. Cell. Cardiol. 52:520–525; 2012. [396] Yu, S. S.; Cai, Y.; Ye, J. T.; Pi, R. B.; Chen, S. R.; Liu, P. Q.; Shen, X. Y.; Ji, Y. Sirtuin 6 protects cardiomyocytes from hypertrophy in vitro via inhibition of NF-kB-dependent transcriptional activity. Br. J. Pharmacol. ; February 15, 2012. [Epub ahead of print]. [397] Cai, Y.; Yu, S. S.; Chen, S. R.; Pi, R. B.; Gao, S.; Li, H.; Ye, J. T.; Liu, P. Q. Nmnat2 protects cardiomyocytes from hypertrophy via activation of SIRT6. FEBS Lett 586:866–874; 2012. [398] Kume, S.; Uzu, T.; Horiike, K.; Chin-Kanasaki, M.; Isshiki, K.; Araki, S.; Sugimoto, T.; Haneda, M.; Kashiwagi, A.; Koya, D. Calorie restriction enhances cell adaptation to hypoxia through Sirt1-dependent mitochondrial autophagy in mouse aged kidney. J. Clin. Invest. 120:1043–1055; 2010. [399] Hasegawa, K.; Wakino, S.; Yoshioka, K.; Tatematsu, S.; Hara, Y.; Minakuchi, H.; Sueyasu, K.; Washida, N.; Tokuyama, H.; Tzukerman, M.; Skorecki, K.; Hayashi, K.; Itoh, H. Kidney-specific overexpression of Sirt1 protects against acute kidney injury by retaining peroxisome function. J. Biol. Chem. 285:13045–13056; 2010. [400] He, W.; Wang, Y.; Zhang, M. Z.; You, L.; Davis, L. S.; Fan, H.; Yang, H. C.; Fogo, A. B.; Zent, R.; Harris, R. C.; Breyer, M. D.; Hao, C. M. Sirt1 activation protects

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

[401]

[402]

[403]

[404]

[405] [406] [407]

[408]

[409]

[410]

[411]

[412]

[413] [414]

[415]

[416]

[417]

[418]

[419]

[420]

[421]

[422]

[423]

the mouse renal medulla from oxidative injury. J. Clin. Invest. 120: 1056–1068; 2010. Yuan, Y.; Chen, Y.; Zhang, P.; Huang, S.; Zhu, C.; Ding, G.; Liu, B.; Yang, T.; Zhang, A. Mitochondrial dysfunction accounts for aldosterone-induced epithelial-to-mesenchymal transition of renal proximal tubular epithelial cells. Free Radic. Biol. Med. 53:30–43; 2012. Koyama, T.; Kume, S.; Koya, D.; Araki, S.; Isshiki, K.; Chin-Kanasaki, M.; Sugimoto, T.; Haneda, M.; Sugaya, T.; Kashiwagi, A.; Maegawa, H.; Uzu, T. SIRT3 attenuates palmitate-induced ROS production and inflammation in proximal tubular cells. Free Radic. Biol. Med. 51:1258–1267; 2011. ¨ J.; Guan, J. S.; Pan, L.; Mak, G.; Kim, Gao, J.; Wang, W. Y.; Mao, Y. W.; Graff, D.; Su, S. C.; Tsai, L. H. A novel pathway regulates memory and plasticity via SIRT1 and miR-134. Nature 466:1105–1109; 2010. Micha´n, S.; Li, Y.; Chou, M. M.; Parrella, E.; Ge, H.; Long, J. M.; Allard, J. S.; Lewis, K.; Miller, M.; Xu, W.; Mervis, R. F.; Chen, J.; Guerin, K. I.; Smith, L. E.; McBurney, M. W.; Sinclair, D. A.; Baudry, M.; de Cabo, R.; Longo, V. D. SIRT1 is essential for normal cognitive function and synaptic plasticity. J. Neurosci. 30:9695–9707; 2010. Aranha, M. M.; Santos, D. M.; Sola´, S.; Steer, C. J.; Rodrigues, C. M. miR-34a regulates mouse neural stem cell differentiation. PLoS One 6:e21396; 2011. Yamakuchi, M.; Lowenstein, C. J. MiR-34, SIRT1 and p53: the feedback loop. Cell Cycle 8:712–715; 2009. Araki, T.; Sasaki, Y.; Milbrandt, J. Increased nuclear NAD biosynthesis and SIRT1 activation prevent axonal degeneration. Science 305:1010–1013; 2004. Kim, D.; Nguyen, M. D.; Dobbin, M. M.; Fischer, A.; Sananbenesi, F.; Rodgers, J. T.; Delalle, I.; Baur, J. A.; Sui, G.; Armour, S. M.; Puigserver, P.; Sinclair, D. A.; Tsai, L. H. SIRT1 deacetylase protects against neurodegeneration in models for Alzheimer’s disease and amyotrophic lateral sclerosis. EMBO J 26:3169–3179; 2007. Chen, J.; Zhou, Y.; Mueller-Steiner, S.; Chen, L. F.; Kwon, H.; Yi, S.; Mucke, L.; Gan, L. SIRT1 protects against microglia-dependent amyloid-beta toxicity through inhibiting NF-kB signaling. J. Biol. Chem. 280:40364–40374; 2005. Min, S. W.; Cho, S. H.; Zhou, Y.; Schroeder, S.; Haroutunian, V.; Seeley, W. W.; Huang, E. J.; Shen, Y.; Masliah, E.; Mukherjee, C.; Meyers, D.; Cole, P. A.; Ott, M.; Gan, L. Acetylation of tau inhibits its degradation and contributes to tauopathy. Neuron 67:953–966; 2010. Donmez, G.; Wang, D.; Cohen, D. E.; Guarente, L. SIRT1 suppresses betaamyloid production by activating the alpha-secretase gene ADAM10. Cell 142:320–332; 2010. Tippmann, F.; Hundt, J.; Schneider, A.; Endres, K.; Fahrenholz, F. Upregulation of the alpha-secretase ADAM10 by retinoic acid receptors and acitretin. FASEB J 23:1643–1654; 2009. Costa, R. M.; Drew, C.; Silva, A. J. Notch to remember. Trends Neurosci. 28:429–435; 2005. Qin, W.; Yang, T.; Ho, L.; Zhao, Z.; Wang, J.; Chen, L.; Zhao, W.; Thiyagarajan, M.; MacGrogan, D.; Rodgers, J. T.; Puigserver, P.; Sadoshima, J.; Deng, H.; Pedrini, S.; Gandy, S.; Sauve, A. A.; Pasinetti, G. M. Neuronal SIRT1 activation as a novel mechanism underlying the prevention of Alzheimer disease amyloid neuropathology by calorie restriction. J. Biol. Chem. 281:21745–21754; 2006. Guo, W.; Qian, L.; Zhang, J.; Zhang, W.; Morrison, A.; Hayes, P.; Wilson, S.; Chen, T.; Zhao, J. Sirt1 overexpression in neurons promotes neurite outgrowth and cell survival through inhibition of the mTOR signaling. J. Neurosci. Res. 89:1723–1736; 2011. Greco, S. J.; Hamzelou, A.; Johnston, J. M.; Smith, M. A.; Ashford, J. W.; Tezapsidis, N. Leptin boosts cellular metabolism by activating AMPK and the sirtuins to reduce tau phosphorylation and b-amyloid in neurons. Biochem. Biophys. Res. Commun. 414:170–174; 2011. Cullen, K. M.; Ko´csi, Z.; Stone, J. Microvascular pathology in the aging human brain: evidence that senile plaques are sites of microhaemorrhages. Neurobiol. Aging 27:1786–1796; 2006. Bell, R. D.; Winkler, E. A.; Singh, I.; Sagare, A. P.; Deane, R.; Wu, Z.; Holtzman, D. M.; Betsholtz, C.; Armulik, A.; Sallstrom, J.; Berk, B. C.; Zlokovic, B. V. Apolipoprotein E controls cerebrovascular integrity via cyclophilin A. Nature 485:512–516; 2012. Bonda, D. J.; Lee, H. G.; Camins, A.; Palla s, M.; Casadesus, G.; Smith, M. A.; Zhu, X. The sirtuin pathway in ageing and Alzheimer disease: mechanistic and therapeutic considerations. Lancet Neurol. 10:275–279; 2012. Korner, S.; Boselt, S.; Thau, N.; Rath, K. J.; Dengler, R.; Petri, S. Differential sirtuin expression patterns in amyotrophic lateral sclerosis (ALS) postmortem tissue: neuroprotective or neurotoxic properties of sirtuins in ALS? Neurodegener. Dis. ; July 10, 2012. [Epub ahead of print]. Lee, J. C.; Shin, J. H.; Park, B. W.; Kim, G. S.; Kim, J. C.; Kang, K. S.; Cha, C. I. Region-specific changes in the immunoreactivity of SIRT1 expression in the central nervous system of SOD1(G93A) transgenic mice as an in vivo model of amyotrophic lateral sclerosis. Brain Res. 1433:20–28; 2012. Wang, J.; Zhang, Y.; Tang, L.; Zhang, N.; Fan, D. Protective effects of resveratrol through the up-regulation of SIRT1 expression in the mutant hSOD1-G93A-bearing motor neuron-like cell culture model of amyotrophic lateral sclerosis. Neurosci. Lett 503:250–255; 2011. Jeong, H.; Cohen, D. E.; Cui, L.; Supinski, A.; Savas, J. N.; Mazzulli, J. R.; Yates 3rd J.; Bordone, L.; Guarente, L.; Krainc, D. Sirt1 mediates neuroprotection from mutant huntingtin by activation of the TORC1 and CREB transcriptional pathway. Nat. Med 18:159–165; 2011.

167

[424] Jiang, M.; Wang, J.; Fu, J.; Du, L.; Jeong, H.; West, T.; Xiang, L.; Peng, Q.; Hou, Z.; Cai, H.; Seredenina, T.; Arbez, N.; Zhu, S.; Sommers, K.; Qian, J.; Zhang, J.; Mori, S.; Yang, X. W.; Tamashiro, K. L.; Aja, S.; Moran, T. H.; Luthi-Carter, R.; Martin, B.; Maudsley, S.; Mattson, M. P.; Cichewicz, R. H.; Ross, C. A.; Holtzman, D. M.; Krainc, D.; Duan, W. Neuroprotective role of Sirt1 in mammalian models of Huntington’s disease through activation of multiple Sirt1 targets. Nat. Med 18:153–158; 2011. [425] Wu, Y.; Li, X.; Zhu, J. X.; Xie, W.; Le, W.; Fan, Z.; Jankovic, J.; Pan, T. Resveratrol-activated AMPK/SIRT1/autophagy in cellular models of Parkinson’s disease. Neurosignals 19:163–174; 2011. [426] Donmez, G.; Arun, A.; Chung, C. Y.; McLean, P. J.; Lindquist, S.; Guarente, L. SIRT1 protects against a-synuclein aggregation by activating molecular chaperones. J. Neurosci. 32:124–132; 2012. [427] Jeong, J. K.; Moon, M. H.; Lee, Y. J.; Seol, J. W.; Park, S. Y. Autophagy induced by the class III histone deacetylase Sirt1 prevents prion peptide neurotoxicity. Neurobiol. Aging ; May 8, 2012. [Epub ahead of print]. [428] Yu, Z.; Fan, D.; Gui, B.; Shi, L.; Xuan, C.; Shan, L.; Wang, Q.; Shang, Y.; Wang, Y. Neurodegeneration-associated TDP-43 interacts with Fragile X mental retardation protein (FMRP)/Staufen (STAU1) and regulates SIRT1 expression in neuronal cells. J. Biol. Chem. 287; 2012. 22560-22472. [429] Outeiro, T. F.; Kontopoulos, E.; Altmann, S. M.; Kufareva, I.; Strathearn, K. E.; Amore, A. M.; Volk, C. B.; Maxwell, M. M.; Rochet, J. C.; McLean, P. J.; Young, A. B.; Abagyan, R.; Feany, M. B.; Hyman, B. T.; Kazantsev, A. G. Sirtuin 2 inhibitors rescue alpha-synuclein-mediated toxicity in models of Parkinson’s disease. Science 317:516–519; 2007. [430] Liu, L.; Arun, A.; Ellis, L.; Peritore, C.; Donmez, G. Sirtuin 2 (SIRT2) enhances 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced nigrostriatal damage via deacetylating Forkhead box O3a (Foxo3a) and activating Bim. J. Biol. Chem 287:32307–32311; 2012. [431] Pallos, J.; Bodai, L.; Lukacsovich, T.; Purcell, J. M.; Steffan, J. S.; Thompson, L. M.; Marsh, J. L. Inhibition of specific HDACs and sirtuins suppresses pathogenesis in a Drosophila model of Huntington’s disease. Hum. Mol. Genet 17:3767–3775; 2008. [432] Luthi-Carter, R.; Taylor, D. M.; Pallos, J.; Lambert, E.; Amore, A.; Parker, A.; Moffitt, H.; Smith, D. L.; Runne, H.; Gokce, O.; Kuhn, A.; Xiang, Z.; Maxwell, M. M.; Reeves, S. A.; Bates, G. P.; Neri, C.; Thompson, L. M.; Marsh, J. L.; Kazantsev, A. G. SIRT2 inhibition achieves neuroprotection by decreasing sterol biosynthesis. Proc. Natl. Acad. Sci. USA 107:7927–7932; 2010. [433] Taylor, D. M.; Balabadra, U.; Xiang, Z.; Woodman, B.; Meade, S.; Amore, A.; Maxwell, M. M.; Reeves, S.; Bates, G. P.; Luthi-Carter, R.; Lowden, P. A.; Kazantsev, A. G. A brain-permeable small molecule reduces neuronal cholesterol by inhibiting activity of sirtuin 2 deacetylase. ACS Chem. Biol. 6:540–546; 2011. [434] Gal, J.; Bang, Y.; Choi, H. J. SIRT2 interferes with autophagy-mediated degradation of protein aggregates in neuronal cells under proteasome inhibition. Neurochem. Int ; July 20, 2012. [Epub ahead of print]. [435] Bosch-Presegue´, L.; Vaquero, A. The dual role of sirtuins in cancer. Genes Cancer 2:648–662; 2011. [436] Deng, C. X. SIRT1, is it a tumor promoter or tumor suppressor? Int. J. Biol. Sci. 5:147–152; 2009. [437] Liu, T.; Liu, P. Y.; Marshall, G. M. The critical role of the class III histone deacetylase SIRT1 in cancer. Cancer Res. 69:1702–1705; 2009. [438] Lee, H.; Kim, K. R.; Noh, S. J.; Park, H. S.; Kwon, K. S.; Park, B. H.; Jung, S. H.; Youn, H. J.; Lee, B. K.; Chung, M. J.; Koh, D. H.; Moon, W. S.; Jang, K. Y. Expression of DBC1 and SIRT1 is associated with poor prognosis for breast carcinoma. Hum. Pathol. 42:204–212; 2011. [439] Huffman, D. M.; Grizzle, W. E.; Bamman, M. M.; Kim, J. S.; Eltoum, I. A.; Elgavish, A.; Nagy, T. R. SIRT1 is significantly elevated in mouse and human prostate cancer. Cancer Res. 67:6612–6618; 2007. ¨ [440] Stunkel, W.; Peh, B. K.; Tan, Y. C.; Nayagam, V. M.; Wang, X.; Salto-Tellez, M.; Ni, B.; Entzeroth, M.; Wood, J. Function of the SIRT1 protein deacetylase in cancer. Biotechnol. J 2:1360–1368; 2007. [441] Chen, J.; Zhang, B.; Wong, N.; Lo, A. W.; To, K. F.; Chan, A. W.; Ng, M. H.; Ho, C. Y.; Cheng, S. H.; Lai, P. B.; Yu, J.; Ng, H. K.; Ling, M. T.; Huang, A. L.; Cai, X. F.; Ko, B. C. Sirtuin 1 is upregulated in a subset of hepatocellular carcinomas where it is essential for telomere maintenance and tumor cell growth. Cancer Res. 71:4138–4149; 2011. [442] Choi, H. N.; Bae, J. S.; Jamiyandorj, U.; Noh, S. J.; Park, H. S.; Jang, K. Y.; Chung, M. J.; Kang, M. J.; Lee, D. G.; Moon, W. S. Expression and role of SIRT1 in hepatocellular carcinoma. Oncol. Rep. 26:503–510; 2012. [443] Zhao, G.; Cui, J.; Zhang, J. G.; Qin, Q.; Chen, Q.; Yin, T.; Deng, S. C.; Liu, Y.; Liu, L.; Wang, B.; Tian, K.; Wang, G. B.; Wang, C. Y. SIRT1 RNAi knockdown induces apoptosis and senescence, inhibits invasion and enhances chemosensitivity in pancreatic cancer cells. Gene Ther 18:920–928; 2011. [444] Hida, Y.; Kubo, Y.; Murao, K.; Arase, S. Strong expression of a longevity-related protein, SIRT1, in Bowen’s disease. Arch. Dermatol. Res. 299:103–106; 2007. [445] Benavente, C. A.; Schnell, S. A.; Jacobson, E. L. Effects of niacin restriction on sirtuin and PARP responses to photodamage in human skin. PLoS One 7:e42276; 2012. [446] Bradbury, C. A.; Khanim, F. L.; Hayden, R.; Bunce, C. M.; White, D. A.; Drayson, M. T.; Craddock, C.; Turner, B. M. Histone deacetylases in acute myeloid leukaemia show a distinctive pattern of expression that changes selectively in response to deacetylase inhibitors. Leukemia 19:1751–1759; 2005. [447] Kozako, T.; Aikawa, A.; Shoji, T.; Fujimoto, T.; Yoshimitsu, M.; Shirasawa, S.; Tanaka, H.; Honda, S. I.; Shimeno, H.; Arima, N.; Soeda, S. High expression of

168

[448] [449]

[450]

[451]

[452]

[453]

[454] [455]

[456]

[457]

[458]

[459]

[460]

[461] [462]

[463]

[464] [465]

[466]

[467]

[468] [469]

[470]

[471]

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

the longevity gene product SIRT1 and apoptosis induction by sirtinol in adult T-cell leukemia cells. Int. J. Cancer 131:2044–2055; 2012. Lim, C. S. SIRT1: tumor promoter or tumor suppressor? Med. Hypotheses 67:341–344; 2006. Fraga, M. F.; Ballestar, E.; Villar-Garea, A.; Boix-Chornet, M.; Espada, J.; Schotta, G.; Bonaldi, T.; Haydon, C.; Ropero, S.; Petrie, K.; Iyer, N. G.; Pe´rezRosado, A.; Calvo, E.; Lopez, J. A.; Cano, A.; Calasanz, M. J.; Colomer, D.; Piris, M. A.; Ahn, N.; Imhof, A.; Caldas, C.; Jenuwein, T.; Esteller, M. Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer. Nat. Genet. 37:391–400; 2005. Pruitt, K.; Zinn, R. L.; Ohm, J. E.; McGarvey, K. M.; Kang, S. H.; Watkins, D. N.; Herman, J. G.; Baylin, S. B. Inhibition of SIRT1 reactivates silenced cancer genes without loss of promoter DNA hypermethylation. PLoS Genet. 2:e40; 2006. Suzuki, K.; Hayashi, R.; Ichikawa, T.; Imanishi, S.; Yamada, T.; Inomata, M.; Miwa, T.; Matsui, S.; Usui, I.; Urakaze, M.; Matsuya, Y.; Ogawa, H.; Sakurai, H.; Saiki, I.; Tobe, K. SRT1720, a SIRT1 activator, promotes tumor cell migration, and lung metastasis of breast cancer in mice. Oncol. Rep. 27:1726–1732; 2012. Caballero, R.; Setien, F.; Lopez-Serra, L.; Boix-Chornet, M.; Fraga, M. F.; Ropero, S.; Megias, D.; Alaminos, M.; Sanchez-Tapia, E. M.; Montoya, M. C.; Esteller, M.; Gonzalez-Sarmiento, R.; Ballestar, E. Combinatorial effects of splice variants modulate function of Aiolos. J. Cell Sci. 120:2619–2630; 2007. Luo, J.; Nikolaev, A. Y.; Imai, S.; Chen, D.; Su, F.; Shiloh, A.; Guarente, L.; Gu, W. Negative control of p53 by Sir2a promotes cell survival under stress. Cell 107:137–148; 2001. van Leeuwen, I.; Lain, S. Sirtuins and p53. Adv. Cancer Res. 102:171–195; 2007. Li, L.; Wang, L.; Li, L.; Wang, Z.; Ho, Y.; McDonald, T.; Holyoake, T. L.; Chen, W.; Bhatia, R. Activation of p53 by SIRT1 inhibition enhances elimination of CML leukemia stem cells in combination with imatinib. Cancer Cell 21:266–281; 2012. Yuan, H.; Wang, Z.; Li, L.; Zhang, H.; Modi, H.; Horne, D.; Bhatia, R.; Chen, W. Activation of stress response gene SIRT1 by BCR-ABL promotes leukemogenesis. Blood 119:1904–1914; 2012. Leko, V.; Varnum-Finney, B.; Li, H.; Gu, Y.; Flowers, D.; Nourigat, C.; Bernstein, I. D.; Bedalov, A. SIRT1 is dispensable for function of hematopoietic stem cells in adult mice. Blood 119:1856–1860; 2012. Dixit, D.; Sharma, V.; Ghosh, S.; Mehta, V. S.; Sen, E. Inhibition of casein kinase-2 induces p53-dependent cell cycle arrest and sensitizes glioblastoma cells to tumor necrosis factor (TNFa)-induced apoptosis through SIRT1 inhibition. Cell Death Dis 3:e271; 2012. Lynch, C. J.; Shah, Z. H.; Allison, S. J.; Ahmed, S. U.; Ford, J.; Warnock, L. J.; Li, H.; Serrano, M.; Milner, J. SIRT1 undergoes alternative splicing in a novel auto-regulatory loop with p53. PLoS One 5:e13502; 2012. Shah, Z. H.; Ahmed, S. U.; Ford, J. R.; Allison, S. J.; Knight, J. R.; Milner, J. A deacetylase-deficient SIRT1 variant opposes full-length SIRT1 in regulating tumor suppressor p53 and governs expression of cancer-related genes. Mol. Cell. Biol. 32:704–716; 2012. Kamel, C.; Abrol, M.; Jardine, K.; He, X.; McBurney, M. W. SirT1 fails to affect p53-mediated biological functions. Aging Cell 5:81–88; 2006. Li, Y.; Matsumori, H.; Nakayama, Y.; Osaki, M.; Kojima, H.; Kurimasa, A.; Ito, H.; Mori, S.; Katoh, M.; Oshimura, M.; Inoue, T. SIRT2 down-regulation in HeLa can induce p53 accumulation via p38 MAPK activation-dependent p300 decrease, eventually leading to apoptosis. Genes Cells 16:34–45; 2011. Li, S.; Banck, M.; Mujtaba, S.; Zhou, M. M.; Sugrue, M. M.; Walsh, M. J. p53induced growth arrest is regulated by the mitochondrial SirT3 deacetylase. PLoS One 5:e10486; 2010. Wong, S.; Weber, J. D. Deacetylation of the retinoblastoma tumour suppressor protein by SIRT1. Biochem. J. 407:451–460; 2007. Heltweg, B.; Gatbonton, T.; Schuler, A. D.; Posakony, J.; Li, H.; Goehle, S.; Kollipara, R.; Depinho, R. A.; Gu, Y.; Simon, J. A.; Bedalov, A. Antitumor activity of a small-molecule inhibitor of human silent information regulator 2 enzymes. Cancer Res. 66:4368–4377; 2006. Yuasa, H.; Oike, Y.; Iwama, A.; Nishikata, I.; Sugiyama, D.; Perkins, A.; Mucenski, M. L.; Suda, T.; Morishita, K. Oncogenic transcription factor Evi1 regulates hematopoietic stem cell proliferation through GATA-2 expression. EMBO J 24:1976–1987; 2005. Pradhan, A. K.; Kuila, N.; Singh, S.; Chakraborty, S. EVI1 up-regulates the stress responsive gene SIRT1 which triggers deacetylation and degradation of EVI1. Biochim. Biophys. Acta 269-275:2011; 1809. Yuan, J.; Minter-Dykhouse, K.; Lou, Z. A c-Myc-SIRT1 feedback loop regulates cell growth and transformation. J. Cell Biol. 185:203–211; 2009. Marshall, G. M.; Liu, P. Y.; Gherardi, S.; Scarlett, C. J.; Bedalov, A.; Xu, N.; Iraci, N.; Valli, E.; Ling, D.; Thomas, W.; van Bekkum, M.; Sekyere, E.; Jankowski, K.; Trahair, T.; Mackenzie, K. L.; Haber, M.; Norris, M. D.; Biankin, A. V.; Perini, G.; Liu, T. SIRT1 promotes N-Myc oncogenesis through a positive feedback loop involving the effects of MKP3 and ERK on N-Myc protein stability. PLoS Genet. 7:e1002135; 2012. Annabi, B.; Lord-Dufour, S.; Ve´zina, A.; Be´liveau, R. Resveratrol targeting of carcinogen-induced brain endothelial cell inflammation biomarkers MMP-9 and COX-2 is Sirt1-independent. Drug Target Insights 6:1–11; 2012. ¨ Menssen, A.; Hydbring, P.; Kapelle, K.; Vervoorts, J.; Diebold, J.; Luscher, B.; Larsson, L. G.; Hermeking, H. The c-MYC oncoprotein, the NAMPT enzyme, the SIRT1-inhibitor DBC1, and the SIRT1 deacetylase form a positive feedback loop. Proc. Natl. Acad. Sci. USA 109:E187–E196; 2012.

[472] Yamashita, S.; Ogawa, K.; Ikei, T.; Udono, M.; Fujiki, T.; Katakura, Y. SIRT1 prevents replicative senescence of normal human umbilical cord fibroblast through potentiating the transcription of human telomerase reverse transcriptase gene. Biochem. Biophys. Res. Commun. 417:630–634; 2012. [473] Pazienza, V.; Piepoli, A.; Panza, A.; Valvano, M. R.; Benegiamo, G.; Vinciguerra, M.; Andriulli, A.; Mazzoccoli, G. SIRT1 and the clock gene machinery in colorectal cancer. Cancer Invest. 30:98–105; 2012. [474] Cha, E. J.; Noh, S. J.; Kwon, K. S.; Kim, C. Y.; Park, B. H.; Park, H. S.; Lee, H.; Chung, M. J.; Kang, M. J.; Lee, D. G.; Moon, W. S.; Jang, K. Y. Expression of DBC1 and SIRT1 is associated with poor prognosis of gastric carcinoma. Clin. Cancer Res. 15:4453–4459; 2009. [475] Sung, J. Y.; Kim, R.; Kim, J. E.; Lee, J. Balance between SIRT1 and DBC1 expression is lost in breast cancer. Cancer Sci. 101:1738–1744; 2010. [476] Feng, A. N.; Zhang, L. H.; Fan, X. S.; Huang, Q.; Ye, Q.; Wu, H. Y.; Yang, J. Expression of SIRT1 in gastric cardiac [sic!] cancer and its clinicopathologic significance. Int. J. Surg. Pathol. 19:743–750; 2011. [477] Zhu, H.; Xia, L.; Zhang, Y.; Wang, H.; Xu, W.; Hu, H.; Wang, J.; Xin, J.; Gang, Y.; Sha, S.; Xu, B.; Fan, D.; Nie, Y.; Wu, K. Activating transcription factor 4 confers a multidrug resistance phenotype to gastric cancer cells through transactivation of SIRT1 expression. PLoS One 7:e31431; 2012. [478] Wang, H.; Liu, H.; Chen, K.; Xiao, J.; He, K.; Zhang, J.; Xiang, G. SIRT1 promotes tumorigenesis of hepatocellular carcinoma through PI3K/PTEN/ AKT signaling. Oncol. Rep. 28:311–318; 2012. [479] Peng, L.; Yuan, Z.; Ling, H.; Fukasawa, K.; Robertson, K.; Olashaw, N.; Koomen, J.; Chen, J.; Lane, W. S.; Seto, E. SIRT1 deacetylates the DNA methyltransferase 1 (DNMT1) protein and alters its activities. Mol. Cell. Biol. 31:4720–4734; 2011. [480] O’Hagan, H. M.; Wang, W.; Sen, S.; Destefano Shields, C.; Lee, S. S.; Zhang, Y. W.; Clements, E. G.; Cai, Y.; Van Neste, L.; Easwaran, H.; Casero, R. A.; Sears, C. L.; Baylin, S. B. Oxidative damage targets complexes containing DNA methyltransferases, SIRT1, and polycomb members to promoter CpG Islands. Cancer Cell 20:606–619; 2012. [481] Benayoun, B. A.; Georges, A. B.; L’Hˆote, D.; Andersson, N.; Dipietromaria, A.; Todeschini, A. L.; Caburet, S.; Bazin, C.; Anttonen, M.; Veitia, R. A. Transcription factor FOXL2 protects granulosa cells from stress and delays cell cycle: role of its regulation by the SIRT1 deacetylase. Hum. Mol. Genet 20:1673–1686; 2011. [482] Welch, C.; Chen, Y.; Stallings, R. L. MicroRNA-34a functions as a potential tumor suppressor by inducing apoptosis in neuroblastoma cells. Oncogene 26:5017–5022; 2007. [483] Akao, Y.; Noguchi, S.; Iio, A.; Kojima, K.; Takagi, T.; Naoe, T. Dysregulation of microRNA-34a expression causes drug-resistance to 5-FU in human colon cancer DLD-1 cells. Cancer Lett. 300:197–204; 2011. [484] Liu, P.; Wilson, M. J. miR-520c and miR-373 upregulate MMP9 expression by targeting mTOR and SIRT1, and activate the Ras/Raf/MEK/Erk signaling pathway and NF-kB factor in human fibrosarcoma cells. J. Cell. Physiol 227:867–876; 2012. [485] Eades, G.; Yao, Y.; Yang, M.; Zhang, Y.; Chumsri, S.; Zhou, Q. miR-200a regulates SIRT1 expression and epithelial to mesenchymal transition (EMT)-like transformation in mammary epithelial cells. J. Biol. Chem. 286:25992–26002; 2011. [486] Lages, E.; Guttin, A.; El Atifi, M.; Ramus, C.; Ipas, H.; Dupre´, I.; Rolland, D.; Salon, C.; Godfraind, C.; deFraipont, F.; Dhobb, M.; Pelletier, L.; Wion, D.; Gay, E.; Berger, F.; Issartel, J. MicroRNA and target protein patterns reveal physiopathological features of glioma subtypes. PLoS One 6:e20600; 2011. [487] Cea, M.; Soncini, D.; Fruscione, F.; Raffaghello, L.; Garuti, A.; Emionite, L.; Moran, E.; Magnone, M.; Zoppoli, G.; Reverberi, D.; Caffa, I.; Salis, A.; Cagnetta, A.; Bergamaschi, M.; Casciaro, S.; Pierri, I.; Damonte, G.; Ansaldi, F.; Gobbi, M.; Pistoia, V.; Ballestrero, A.; Patrone, F.; Bruzzone, S.; Nencioni, A. Synergistic interactions between HDAC and sirtuin inhibitors in human leukemia cells. PLoS One 6:e22739; 2011. [488] Kojima, K.; Ohhashi, R.; Fujita, Y.; Hamada, N.; Akao, Y.; Nozawa, Y.; Deguchi, T.; Ito, M. A role for SIRT1 in cell growth and chemoresistance in prostate cancer PC3 and DU145 cells. Biochem. Biophys. Res. Commun. 373:423–428; 2008. [489] Wang, T. T. Y.; Schoene, N. W.; Kim, E. -K.; Kim, Y. S. Pleiotropic effects of the sirtuin inhibitor sirtinol involves concentration-dependent modulation of multiple nuclear receptor-mediated pathways in androgen-responsive prostate cancer cell LNCaP. Mol. Carcinog ; April 11, 2012. [Epub ahead of print]. [490] Nakane, K.; Fujita, Y.; Terazawa, R.; Atsumi, Y.; Kato, T.; Nozawa, Y.; Deguchi, T.; Ito, M. Inhibition of cortactin and SIRT1 expression attenuates migration and invasion of prostate cancer DU145 cells. Int. J. Urol. 19:71–79; 2012. [491] Wang, B.; Hasan, M. K.; Alvarado, E.; Yuan, H.; Wu, H.; Chen, W. Y. NAMPT overexpression in prostate cancer and its contribution to tumor cell survival and stress response. Oncogene 30:907–921; 2011. [492] Byles, V.; Chmilewski, L. K.; Wang, J.; Zhu, L.; Forman, L. W.; Faller, D. V.; Dai, Y. Aberrant cytoplasm localization and protein stability of SIRT1 is regulated by PI3K/IGF-1R signaling in human cancer cells. Int. J. Biol. Sci. 6:599–612; 2010. [493] Oberdoerffer, P.; Michan, S.; McVay, M.; Mostoslavsky, R.; Vann, J.; Park, S. K.; Hartlerode, A.; Stegmuller, J.; Hafner, A.; Loerch, P.; Wright, S. M.; Mills, K. D.; Bonni, A.; Yankner, B. A.; Scully, R.; Prolla, T. A.; Alt, F. W.; Sinclair, D. A. SIRT1 redistribution on chromatin promotes genomic stability but alters gene expression during aging. Cell 135:907–918; 2008. [494] Firestein, R.; Blander, G.; Michan, S.; Oberdoerffer, P.; Ogino, S.; Campbell, J.; Bhimavarapu, A.; Luikenhuis, S.; de Cabo, R.; Fuchs, C.; Hahn, W. C.;

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

[495]

[496]

[497]

[498]

[499]

[500] [501]

[502]

[503]

[504]

[505]

[506]

[507]

[508]

[509]

[510] [511]

[512]

[513]

[514]

[515]

Guarente, D. A.; Sinclair, D. A. The SIRT1 deacetylase suppresses intestinal tumorigenesis and colon cancer growth. PLoS One 3:e2020; 2008. Wang, R. H.; Zheng, Y.; Kim, H. S.; Xu, X.; Cao, L.; Luhasen, T.; Lee, M. H.; Xiao, C.; Vassilopoulos, A.; Chen, W.; Gardner, K.; Man, Y. G.; Hung, M. C.; Finkel, T.; Deng, C. X. Interplay among BRCA1, SIRT1, and Survivin during BRCA1-associated tumorigenesis. Mol. Cell 32:11–20; 2008. Cho, I. R.; Koh, S. S.; Malilas, W.; Srisuttee, R.; Moon, J.; Choi, Y. W.; Horio, Y.; Oh, S.; Chung, Y. H. SIRT1 inhibits proliferation of pancreatic cancer cells expressing pancreatic adenocarcinoma up-regulated factor (PAUF), a novel oncogene, by suppression of b-catenin. Biochem. Biophys. Res. Commun. 423:270–275; 2012. Singh, N. P.; Singh, U. P.; Hegde, V. L.; Guan, H.; Hofseth, L.; Nagarkatti, M.; Nagarkatti, P. S. Resveratrol (trans-3,5,40 -trihydroxystilbene) suppresses EL4 tumor growth by induction of apoptosis involving reciprocal regulation of SIRT1 and NF-kB. Mol. Nutr. Food Res. 55:1207–1218; 2011. Frazzi, R.; Valli, R.; Tamagnini, I.; Casali, B.; Latruffe, N.; Merli, F. Resveratrol-mediated apoptosis of hodgkin lymphoma cells involves SIRT1 inhibition and FOXO3a hyperacetylation. Int. J. Cancer ; July 26, 2012. [Epub ahead of print]. Zhang, Q.; Zeng, S. X.; Zhang, Y.; Zhang, Y.; Ding, D.; Ye, Q.; Meroueh, S. O.; Lu, H. A small molecule Inauhzin inhibits SIRT1 activity and suppresses tumour growth through activation of p53. EMBO Mol. Med. 4:298–312; 2012. ˜ oz-Martı´n, M.; Serrano, M. Limited role of Sirt1 Herranz, D.; Iglesias, G.; Mun in cancer protection by dietary restriction. Cell Cycle 10:2215–2217; 2011. Murray-Zmijewski, F.; Slee, E. A.; Lu, X. A complex barcode underlies the heterogeneous response of p53 to stress. Nat. Rev. Mol. Cell. Biol. 9:702–712; 2008. ¨ Sahin, E.; Colla, S.; Liesa, M.; Moslehi, J.; Muller, F. L.; Guo, M.; Cooper, M.; Kotton, D.; Fabian, A. J.; Walkey, C.; Maserm, R. S.; Tonon, G.; Foerster, F.; Xiong, R.; Wang, Y. A.; Shukla, S. A.; Jaskelioff, M.; Martin, E. S.; Heffernan, T. P.; Protopopov, A.; Ivanova, E.; Mahoney, J. E.; Kost-Alimova, M.; Perry, S. R.; Bronson, R.; Liao, R.; Mulligan, R.; Shirihai, O. S.; Chin, L.; DePinho, R. A. Telomere dysfunction induces metabolic and mitochondrial compromise. Nature 470:359–365; 2011. Srisuttee, R.; Koh, S. S.; Kim, S. J.; Malilas, W.; Boonying, W.; Cho, I. R.; Jhun, B. H.; Ito, M.; Horio, Y.; Seto, E.; Oh, S.; Chung, Y. H. Hepatitis B virus X (HBX) protein upregulates b-catenin in a human hepatic cell line by sequestering SIRT1 deacetylase. Oncol. Rep. 28:276–282; 2012. Wang, S.; Song, P.; Zou, M. H. Inhibition of AMP-activated protein kinase a (AMPKa) by doxorubicin accentuates genotoxic stress and cell death in mouse embryonic fibroblasts and cardiomyocytes: role of p53 and SIRT1. J. Biol. Chem 287:8001–8012; 2012. Sza´nto´, M.; Rutkai, I.; Hegedus, C.; Czikora, A´.; Ro´zsahegyi, M.; Kiss, B.; Vira´g, L.; Gergely, P.; To´th, A.; Bai, P. Poly(ADP-ribose) polymerase-2 depletion reduces doxorubicin-induced damage through SIRT1 induction. Cardiovasc. Res. 92:430–438; 2011. Andersen, J. L.; Thompson, J. W.; Lindblom, K. R.; Johnson, E. S.; Yang, C. S.; Lilley, L. R.; Freel, C. D.; Moseley, M. A.; Kornbluth, S. A biotin switch-based proteomics approach identifies 14-3-3z as a target of Sirt1 in the metabolic regulation of caspase-2. Mol. Cell 43:834–842; 2011. Jung, Y. J.; Lee, J. E.; Lee, A. S.; Kang, K. P.; Lee, S.; Park, S. K.; Lee, S. Y.; Han, M. K.; Kim, D. H.; Kim, W. SIRT1 overexpression decreases cisplatin-induced acetylation of NF-kB p65 subunit and cytotoxicity in renal proximal tubule cells. Biochem. Biophys. Res. Commun. 419:206–210; 2012. Inoue, T.; Hiratsuka, M.; Osaki, M.; Yamada, H.; Kishimoto, I.; Yamaguchi, S.; Nakano, S.; Katoh, M.; Ito, H.; Oshimura, M. SIRT2, a tubulin deacetylase, acts to block the entry to chromosome condensation in response to mitotic stress. Oncogene 26:945–957; 2007. Hiratsuka, M.; Inoue, T.; Toda, T.; Kimura, N.; Shirayoshi, Y.; Kamitani, H.; Watanabe, T.; Ohama, E.; Tahimic, C. G.; Kurimasa, A.; Oshimura, M. Proteomics-based identification of differentially expressed genes in human gliomas: down-regulation of SIRT2 gene. Biochem. Biophys. Res. Commun. 309:558–566; 2003. North, B. J.; Verdin, E. Mitotic regulation of SIRT2 by cyclin-dependent kinase 1-dependent phosphorylation. J. Biol. Chem. 282:19546–19555; 2007. He, X.; Nie, H.; Hong, Y.; Sheng, C.; Xia, W.; Ying, W. SIRT2 activity is required for the survival of C6 glioma cells. Biochem. Biophys. Res. Commun. 417:468–472; 2012. Peters, C. J.; Rees, J. R.; Hardwick, R. H.; Hardwick, J. S.; Vowler, S. L.; Ong, C. A.; Zhang, C.; Save, V.; O’Donovan, M.; Rassl, D.; Alderson, D.; Caldas, C. A 4-gene signature predicts survival of patients with resected adenocarcinoma of the esophagus, junction, and gastric cardia. Gastroenterology 139:1995–2004; 2010. ¨ Lennerz, V.; Fatho, M.; Gentilini, C.; Frye, R. A.; Lifke, A.; Ferel, D.; Wolfel, C.; ¨ Huber, C.; Wolfel, T. The response of autologous T cells to a human melanoma is dominated by mutated neoantigens. Proc. Natl. Acad. Sci. USA 102:16013–16018; 2005. Kim, H. S.; Vassilopoulos, A.; Wang, R. H.; Lahusen, T.; Xiao, Z.; Xu, X.; Li, C.; Veenstra, T. D.; Li, B.; Yu, H.; Ji, J.; Wang, X. W.; Park, S. H.; Cha, Y. I.; Gius, D.; Deng, C. X. SIRT2 maintains genome integrity and suppresses tumorigenesis through regulating APC/C activity. Cancer Cell 20:487–499; 2011. Dan, L.; Klimenkova, O.; Klimiankou, M.; Klusman, J. H.; van den HeuvelEibrink, M. M.; Reinhardt, D.; Welte, K.; Skokowa, J. The role of sirtuin 2 activation by nicotinamide phosphoribosyltransferase in the aberrant proliferation and survival of myeloid leukemia cells. Haematologica 97:551–559; 2011.

169

[516] Alhazzazi, T. Y.; Kamarajan, P.; Verdin, E.; Kapila, Y. L. SIRT3 and cancer: tumor promoter or suppressor? Biochim Biophys Acta. 80-88:2011; 1816. [517] Ashraf, N.; Zino, S.; Macintyre, A.; Kingsmore, D.; Payne, A. P.; George, W. D.; Shiels, P. G. Altered sirtuin expression is associated with node-positive breast cancer. Br. J. Cancer 95:1056–1061; 2007. [518] Zhang, Y. Y.; Zhou, L. M. Sirt3 inhibits hepatocellular carcinoma cell growth through reducing Mdm2-mediated p53 degradation. Biochem. Biophys. Res. Commun. 423:26–31; 2012. [519] Kamarajan, P.; Alhazzazi, T. Y.; Danciu, T.; D’silva, N. J.; Verdin, E.; Kapila, Y. L. Receptor-interacting protein (RIP) and Sirtuin-3 (SIRT3) are on opposite sides of anoikis and tumorigenesis. Cancer ; June 6, 2012. [Epub ahead of print]. [520] Yu, W.; Dittenhafer-Reed, K. E.; Denu, J. M. SIRT3 protein deacetylates isocitrate dehydrogenase 2 (IDH2) and regulates mitochondrial redox status. J. Biol. Chem. 287:14078–14086; 2012. [521] McCord, R. A.; Michishita, E.; Hong, T.; Berber, E.; Boxer, L. D.; Kusumoto, R.; Guan, S.; Shi, X.; Gozani, O.; Burlingame, A. L.; Bohr, V. A.; Chua, K. F. SIRT6 stabilizes DNA-dependent protein kinase at chromatin for DNA doublestrand break repair. Aging (Albany NY) 1:109–121; 2009. [522] Kaidi, A.; Weinert, B. T.; Choudhary, C.; Jackson, S. P. Human SIRT6 promotes DNA end resection through CtIP deacetylation. Science 329:1348–1353; 2010. [523] Van Meter, M.; Mao, Z.; Gorbunova, V.; Seluanov, A. SIRT6 overexpression induces massive apoptosis in cancer cells but not in normal cells. Cell Cycle 10:3153–3158; 2011. [524] Martı´nez-Redondo, P.; Santos-Barriopedro, I.; Vaquero, A. A big step for SIRT7, one giant leap for sirtuinsy in cancer. Cancer Cell 21:719–721; 2012. [525] Chua, K. F.; Mostoslavsky, R.; Lombard, D. B.; Pang, W. W.; Saito, S.; Franco, S.; Kaushal, D.; Cheng, H. L.; Fischer, M. R.; Stokes, N.; Murphy, M. M.; Appella, E.; Alt, F. W. Mammalian SIRT1 limits replicative life span in response to chronic genotoxic stress. Cell Metab. 2:67–76; 2005. [526] Allison, S. J.; Milner, J. SIRT3 is pro-apoptotic and participates in distinct basal apoptotic pathways. Cell Cycle 6:2669–2677; 2007. [527] Langley, E.; Pearson, M.; Faretta, M.; Bauer, U. M.; Frye, R. A.; Minucci, S.; Pelicci, P. G.; Kouzarides, T. Human SIR2 deacetylates p53 and antagonizes PML/p53-induced cellular senescence. EMBO J 21:2383–2396; 2002. [528] Ota, H.; Tokunaga, E.; Chang, K.; Hikasa, M.; Iijima, K.; Eto, M.; Kozaki, K.; Akishita, M.; Ouchi, Y.; Kaneki, M. Sirt1 inhibitor, Sirtinol, induces senescence-like growth arrest with attenuated Ras-MAPK signaling in human cancer cells. Oncogene 25:176–185; 2006. [529] Liu, T. F.; Vachharajani, V. T.; Yoza, B. K.; McCall, C. E. NAD þ -dependent sirtuins 1 and 6 coordinate a switch from glucose to fatty acid oxidation during the acute inflammatory response. J. Biol. Chem. 287:25758–25769; 2012. [530] Gao, B.; Kong, Q.; Kemp, K.; Zhao, Y. S.; Fang, D. Analysis of sirtuin 1 expression reveals a molecular explanation of IL-2-mediated reversal of T-cell tolerance. Proc. Natl. Acad. Sci. USA 109:899–904; 2012. [531] van Loosdregt, J.; Brunen, D.; Fleskens, V.; Pals, C. E.; Lam, E. W.; Coffer, P. J. Rapid temporal control of Foxp3 protein degradation by sirtuin-1. PLoS One 6:e19047; 2011. [532] Zhu, X.; Liu, Q.; Wang, M.; Liang, M.; Yang, X.; Xu, X.; Zou, H.; Qiu, J. Activation of Sirt1 by resveratrol inhibits TNF-a induced inflammation in fibroblasts. PLoS One 6:e27081; 2011. [533] Wu, X.; Kong, X.; Chen, D.; Li, H.; Zhao, Y.; Xia, M.; Fang, M.; Li, P.; Fang, F.; Sun, L.; Tian, W.; Xu, H.; Yang, Y.; Qi, X.; Gao, Y.; Sha, J.; Chen, Q.; Xu, Y. SIRT1 links CIITA deacetylation to MHC II activation. Nucleic Acids Res. 39:9549–9558; 2011. [534] Niederer, F.; Ospelt, C.; Brentano, F.; Hottiger, M. O.; Gay, R. E.; Gay, S.; Detmar, M.; Kyburz, D. SIRT1 overexpression in the rheumatoid arthritis synovium contributes to proinflammatory cytokine production and apoptosis resistance. Ann. Rheum. Dis. 70:1866–1873; 2011. [535] Dvir-Ginzberg, M.; Gagarina, V.; Lee, E. J.; Booth, R.; Gabay, O.; Hall, D. J. Tumor necrosis factor a-mediated cleavage and inactivation of SirT1 in human osteoarthritic chondrocytes. Arthritis Rheum. 63:2363–2373; 2011. [536] Hong, E. H.; Yun, H. S.; Kim, J.; Um, H. D.; Lee, K. H.; Kang, C. M.; Lee, S. J.; Chun, J. S.; Hwang, S. G. Nicotinamide phosphoribosyltransferase is essential for interleukin-1beta-mediated dedifferentiation of articular chondrocytes via SIRT1 and extracellular signal-regulated kinase (ERK) complex signaling. J. Biol. Chem. 286:28619–28631; 2011. [537] Gabay, O.; Oppenhiemer, H.; Meir, H.; Zaal, K.; Sanchez, C.; Dvir-Ginzberg, M. Increased apoptotic chondrocytes in articular cartilage from adult heterozygous SirT1 mice. Ann. Rheum. Dis. 71:613–616; 2012. [538] Lim, H. D.; Kim, Y. S.; Ko, S. H.; Yoon, I. J.; Cho, S. G.; Chun, Y. H.; Choi, B. J.; Kim, E. C. Cytoprotective and anti-inflammatory effects of melatonin in hydrogen peroxide-stimulated CHON-001 human chondrocyte cell line and rabbit model of osteoarthritis via the SIRT1 pathway. J. Pineal Res. 53:225–237; 2012. [539] Lei, M.; Wang, J. G.; Xiao, D. M.; Fan, M.; Wang, D. P.; Xiong, J. Y.; Chen, Y.; Ding, Y.; Liu, S. L. Resveratrol inhibits interleukin 1b-mediated inducible nitric oxide synthase expression in articular chondrocytes by activating SIRT1 and thereby suppressing nuclear factor-kB activity. Eur. J. Pharmacol. 674:73–79; 2012. [540] Tseng, P. C.; Hou, S. M.; Chen, R. J.; Peng, H. W.; Hsieh, C. F.; Kuo, M. L.; Yen, M. L. Resveratrol promotes osteogenesis of human mesenchymal stem cells by upregulating RUNX2 gene expression via the SIRT1/FOXO3A axis. J. Bone Miner. Res. 26:2552–2563; 2011.

170

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

[541] Shakibaei, M.; Shayan, P.; Busch, F.; Aldinger, C.; Buhrmann, C.; Lueders, C.; Mobasheri, A. Resveratrol mediated modulation of sirt-1/runx2 promotes osteogenic differentiation of mesenchymal stem cells: potential role of runx2 deacetylation. PLoS One 7:e35712; 2012. [542] Matsui, K.; Ezoe, S.; Oritani, K.; Shibata, M.; Tokunaga, M.; Fujita, N.; Tanimura, A.; Sudo, T.; Tanaka, H.; McBurney, M. W.; Matsumura, I.; Kanakura, Y. NAD-dependent histone deacetylase, SIRT1, plays essential roles in the maintenance of hematopoietic stem cells. Biochem. Biophys. Res. Commun. 418:811–817; 2012. [543] Cohen-Kfir, E.; Artsi, H.; Levin, A.; Abramowitz, E.; Bajayo, A.; Gurt, I.; Zhong, L.; D’Urso, A.; Toiber, D.; Mostoslavsky, R.; Dresner-Pollak, R. Sirt1 is a regulator of bone mass and a repressor of Sost encoding for sclerostin, a bone formation inhibitor. Endocrinology 152:4514–4524; 2011. [544] Seifert, E. L.; Caron, A. Z.; Morin, K.; Coulombe, J.; He, X. H.; Jardine, K.; Dewar-Darch, D.; Boekelheide, K.; Harper, M. E.; McBurney, M. W. SirT1 catalytic activity is required for male fertility and metabolic homeostasis in mice. FASEB J 26:555–566; 2012. [545] Morita, Y.; Wada-Hiraike, O.; Yano, T.; Shirane, A.; Hirano, M.; Hiraike, H.; Koyama, S.; Oishi, H.; Yoshino, O.; Miyamoto, Y.; Sone, K.; Oda, K.; Nakagawa, S.; Tsutsui, K.; Taketani, Y. Resveratrol promotes expression of SIRT1 and StAR in rat ovarian granulosa cells: an implicative role of SIRT1 in the ovary. Reprod. Biol. Endocrinol. 10:1–10; 2012. [546] Tomada, I.; Tomada, N.; Almeida, H.; Neves, D. Androgen depletion in humans leads to cavernous tissue reorganization and upregulation of Sirt1eNOS axis. Age (Dordr.) ; November 4, 2011. [Epub ahead of print]. [547] Kuroda, S.; Yamazaki, M.; Abe, M.; Sakimura, K.; Takayanagi, H.; Iwai, Y. Basic leucine zipper transcription factor, ATF-like (BATF) regulates epigenetically and energetically effector CD8 T-cell differentiation via Sirt1 expression. Proc. Natl. Acad. Sci. USA 108:14885–14889; 2011. [548] Yao, H.; Chung, S.; Hwang, J. W.; Rajendrasozhan, S.; Sundar, I. K.; Dean, D. ¨ A.; McBurney, M. W.; Guarente, L.; Gu, W.; Ronty, M.; Kinnula, V. L.; Rahman, I. SIRT1 protects against emphysema via FOXO3-mediated reduction of premature senescence in mice. J. Clin. Invest. 122:2032–2045; 2012. [549] Wu, Z.; Liu, M. C.; Liang, M.; Fu, J. Sirt1 protects against thrombomodulin down-regulation and lung coagulation after particulate matter exposure. Blood 119:2422–2429; 2012. [550] Shi, J.; Yin, N.; Xuan, L. L.; Yao, C. S.; Meng, A. M.; Hou, Q. Vam3, a derivative of resveratrol, attenuates cigarette smoke-induced autophagy. Acta Pharmacol. Sin. 33:888–896; 2012. [551] Yin, H.; Hu, M.; Zhang, R.; Shen, Z.; Flatow, L.; You, M. MicroRNA-217 promotes ethanol-induced fat accumulation in hepatocytes by downregulating SIRT1. J. Biol. Chem. 287:9817–9826; 2012. [552] Ajmo, J. M.; Liang, X.; Rogers, C. Q.; Pennock, B.; You, M. Resveratrol alleviates alcoholic fatty liver in mice. Am. J. Physiol. Gastrointest. Liver Physiol. 295:G833–G842; 2008. [553] Fritz, K. S.; Galligan, J. J.; Hirschey, M. D.; Verdin, E.; Petersen, D. R. Mitochondrial acetylome analysis in a mouse model of alcohol-induced liver injury utilizing SIRT3 knockout mice. J. Proteome Res. 11:1633–1643; 2012. [554] Cao, C.; Lu, S.; Kivlin, R.; Wallin, B.; Card, E.; Bagdasarian, A.; Tamakloe, T.; Wang, W. J.; Song, X.; Chu, W. M.; Kouttab, N.; Xu, A.; Wan, Y. SIRT1 confers protection against UVB- and H2O2-induced cell death via modulation of p53 and JNK in cultured skin keratinocytes. J. Cell. Mol. Med. 13:3632–3643; 2009. [555] Momken, I.; Stevens, L.; Bergouignan, A.; Desplanches, D.; Rudwill, F.; Chery, I.; Zahariev, A.; Zahn, S.; Stein, T. P.; Sebedio, J. L.; Pujos-Guillot, E.; Falempin, M.; Simon, C.; Coxam, V.; Andrianjafiniony, T.; Gauquelin-Koch, G.; Picquet, F.; Blanc, S. Resveratrol prevents the wasting disorders of mechanical unloading by acting as a physical exercise mimetic in the rat. FASEB J 25:3646–3660; 2011. [556] Busch, F.; Mobasheri, A.; Shayan, P.; Stahlmann, R.; Shakibaei, M. Sirt-1 is required for the inhibition of apoptosis and inflammatory responses in human tenocytes. J. Biol. Chem. 287:25770–25781; 2012. [557] Balaiya, S.; Ferguson, L. R.; Chalam, K. V. Evaluation of sirtuin role in neuroprotection of retinal ganglion cells in hypoxia. Invest. Ophthalmol. Vis. Sci. 53:4315–4322; 2012. [558] Kourtis, N.; Tavernarakis, N. Cellular stress response pathways and ageing: intricate molecular relationships. EMBO J 30:2520–2531; 2011. [559] Jin, J.; Iakova, P.; Jiang, Y.; Medrano, E. E.; Timchenko, N. A. The reduction of SIRT1 in livers of old mice leads to impaired body homeostasis and to inhibition of liver proliferation. Hepatology 54:989–998; 2011. [560] Qian, S. B.; Zhang, X.; Sun, J.; Bennink, J. R.; Yewdell, J. W.; Patterson, C. mTORC1 links protein quality and quantity control by sensing chaperone availability. J. Biol. Chem. 285:27385–27395; 2010. [561] Chan, S. W.; Blackburn, E. H. New ways not to make ends meet: telomerase, DNA damage proteins and heterochromatin. Oncogene 21:553–563; 2002. [562] El Ramy, R.; Magroun, N.; Messadecq, N.; Gauthier, L. R.; Boussin, F. D.; Kolthur-Seetharam, U.; Schreiber, V.; McBurney, M. W.; Sassone-Corsi, P.; Dantzer, F. Functional interplay between Parp-1 and SirT1 in genome integrity and chromatin-based processes. Cell. Mol. Life Sci. 66:3219–3234; 2009. [563] Palacios, J. A.; Herranz, D.; De Bonis, M. L.; Velasco, S.; Serrano, M.; Blasco, M. A. SIRT1 contributes to telomere maintenance and augments global homologous recombination. J. Cell Biol. 191:1299–1313; 2010. ¨ [564] Sahin, E.; Colla, S.; Liesa, M.; Moslehi, J.; Muller, F. L.; Guo, M.; Cooper, M.; Kotton, D.; Fabian, A. J.; Walkey, C.; Maser, R. S.; Tonon, G.; Foerster, F.; Xiong, R.; Wang, Y. A.; Shukla, S. A.; Jaskelioff, M.; Martin, E. S.; Heffernan, T.

[565]

[566]

[567]

[568]

[569]

[570]

[571]

[572]

[573]

[574]

[575]

[576]

[577] [578]

[579]

[580]

[581]

[582]

[583]

[584]

[585] [586]

[587]

[588]

P.; Protopopov, A.; Ivanova, E.; Mahoney, J. E.; Kost-Alimova, M.; Perry, S. R.; Bronson, R.; Liao, R.; Mulligan, R.; Shirihai, O. S.; Chin, L.; DePinho, R. A. Telomere dysfunction induces metabolic and mitochondrial compromise. Nature 470:359–365; 2011. Krishnakumar, R.; Kraus, W. L. The PARP side of the nucleus: molecular actions, physiological outcomes, and clinical targets. Mol. Cell 39:8–24; 2010. de Murcia, J. M.; Niedergang, C.; Trucco, C.; Ricoul, M.; Dutrillaux, B.; Mark, M.; Oliver, F. J.; Masson, M.; Dierich, A.; LeMeur, M.; Walztinger, C.; Chambon, P.; de Murcia, G. Requirement of poly(ADP-ribose) polymerase in recovery from DNA damage in mice and in cells. Proc. Natl. Acad. Sci. USA 94:7303–7307; 1997. Furukawa, A.; Tada-Oikawa, S.; Kawanishi, S.; Oikawa, S. H2O2 accelerates cellular senescence by accumulation of acetylated p53 via decrease in the function of SIRT1 by NAD þ depletion. Cell Physiol. Biochem 20:45–54; 2007. Me´nissier de Murcia, J.; Ricoul, M.; Tartier, L.; Niedergang, C.; Huber, A.; Dantzer, F.; Schreiber, V.; Ame´, J. C.; Dierich, A.; LeMeur, M.; Sabatier, L.; Chambon, P.; de Murcia, G. Functional interaction between PARP-1 and PARP-2 in chromosome stability and embryonic development in mouse. EMBO J 22:2255–2263; 2003. ¨ Grube, K.; Burkle, A. Poly(ADP-ribose) polymerase activity in mononuclear leukocytes of 13 mammalian species correlates with species-specific life span. Proc. Natl. Acad. Sci. USA 89:11759–11763; 1992. ¨ ¨ ¨ Muiras, M. L.; Muller, M.; Schachter, F.; Burkle, A. Increased poly(ADPribose) polymerase activity in lymphoblastoid cell lines from centenarians. J. Mol. Med. (Berl.) 76:346–354; 1998. Matsushita, H.; Chang, E.; Glassford, A. J.; Cooke, J. P.; Chiu, C. P.; Tsao, P. S. eNOS activity is reduced in senescent human endothelial cells: preservation by hTERT immortalization. Circ. Res. 89:793–798; 2001. Ungvari, Z.; Kaley, G.; de Cabo, R.; Sonntag, W. E.; Csiszar, A. Mechanisms of vascular aging: new perspectives. J. Gerontol. A Biol. Sci. Med. Sci 65:1028–1041; 2010. Menghini, R.; Casagrande, V.; Cardellini, M.; Martelli, E.; Terrinoni, A.; Amati, F.; Vasa-Nicotera, M.; Ippoliti, A.; Novelli, G.; Melino, G.; Lauro, R.; Federici, M. MicroRNA 217 modulates endothelial cell senescence via silent information regulator 1. Circulation 120:1524–1532; 2009. Yuan, H. F.; Zhai, C.; Yan, X. L.; Zhao, D. D.; Wang, J. X.; Zeng, Q.; Chen, L.; Nan, X.; He, L. J.; Li, S. T.; Yue, W.; Pei, X. T. SIRT1 is required for long-term growth of human mesenchymal stem cells. J. Mol. Med. (Berl.) 90:389–400; 2012. Li, Y.; Tollefsbol, T. O. p16INK4a suppression by glucose restriction contributes to human cellular lifespan extension through SIRT1-mediated epigenetic and genetic mechanisms. PLoS One 6:e17421; 2011. Yang, N. C.; Song, T. Y.; Chen, M. Y.; Hu, M. L. Effects of 2-deoxyglucose and dehydroepiandrosterone on intracellular NAD þ level, SIRT1 activity and replicative lifespan of human Hs68 cells. Biogerontology 12:527–536; 2011. Tabata, M.; Pardo, P. S.; Boriek, A. M. The physiological roles of Sirt1 in skeletal muscle. Aging (Albany NY) 3:430–437; 2011. Wenz, T.; Rossi, S. G.; Rotundo, R. L.; Spiegelman, B. M.; Moraes, C. T. Increased muscle PGC-1a expression protects from sarcopenia and metabolic disease during aging. Proc. Natl. Acad. Sci. USA 106:20405–20410; 2009. Borra´s, C.; Monleo´n, D.; Lo´pez-Grueso, R.; Gambini, J.; Orlando, L.; Pallardo´, ˜ a, J.; Font de Mora, J. RasGrf1 deficiency delays aging in F. V.; Santos, E.; Vin mice. Aging (Albany NY) 3:262–276; 2011.  Cristofol, R.; Porquet, D.; Corpas, R.; Coto-Montes, A.; Serret, J.; Camins, A.; Palla s, M.; Sanfeliu, C. Neurons from senescence-accelerated SAMP8 mice are protected against frailty by the sirtuin 1 promoting agents melatonin and resveratrol. J. Pineal Res. 52:271–281; 2012. Ota, H.; Akishita, M.; Akiyoshi, T.; Kahyo, T.; Setou, M.; Ogawa, S.; Iijima, K.; Eto, M.; Ouchi, Y. Testosterone deficiency accelerates neuronal and vascular aging of SAMP8 mice: protective role of eNOS and SIRT1. PLoS One 7:e29598; 2012. Quintas, A.; de Solı´s, A. J.; Dı´ez-Guerra, F. J.; Carrascosa, J. M.; Bogo´nez, E. Age-associated decrease of SIRT1 expression in rat hippocampus: prevention by late onset caloric restriction. Exp. Gerontol. 47:198–201; 2012. Huang, P. S.; Son, J. H.; Abbott, L. C.; Winzer-Serhan, U. H. Regulated expression of neuronal SIRT1 and related genes by aging and neuronal b2containing nicotinic cholinergic receptors. Neuroscience 196:189–202; 2011. Panossian, L.; Fenik, P.; Zhu, Y.; Zhan, G.; McBurney, M. W.; Veasey, S. SIRT1 regulation of wakefulness and senescence-like phenotype in wake neurons. J. Neurosci. 31:4025–4036; 2011. Li, X.; Khanna, A.; Li, N.; Wang, E. Circulatory miR34a as an RNAbased, noninvasive biomarker for brain aging. Aging (Albany NY) 3:985–1002; 2011. Heyn, H.; Li, N.; Ferreira, H. J.; Moran, S.; Pisano, D. G.; Gomez, J.; Diez, A.; Sanchez-Mut, J. V.; Setien, F.; Carmona, F. J.; Puca, A. A.; Sayols, S.; Pujana, M. A.; Serra-Musach, J.; Iglesias-Platas, I.; Formiga, F.; Fernandez, A. F.; Fraga, M. F.; Heath, S. C.; Valencia, A.; Gut, I. G.; Wang, J.; Esteller, M. Distinct DNA methylomes of newborns and centenarians. Proc. Natl. Acad. Sci. USA 109:10522–10527; 2012. Stefanska, B.; Karlic, H.; Varga, F.; Fabianowska-Majewska, K.; Haslberger, A. G. Epigenetic mechanisms in anti-cancer actions of bioactive food components—the implications in cancer prevention. Br. J. Pharmacol. 167:279–297; 2012. Wakeling, L. A.; Ions, L. J.; Ford, D. Could Sirt1-mediated epigenetic effects contribute to the longevity response to dietary restriction and be mimicked by other dietary interventions? Age (Dordr.) 31:327–341; 2009.

B.J. Morris / Free Radical Biology and Medicine 56 (2013) 133–171

[589] Ford, D.; Ions, L. J.; Alatawi, F.; Wakeling, L. A. The potential role of epigenetic responses to diet in ageing. Proc. Nutr. Soc. 70:374–384; 2011. [590] Stefanska, B.; Salame´, P.; Bednarek, A.; Fabianowska-Majewska, K. Comparative effects of retinoic acid, vitamin D and resveratrol alone and in combination with adenosine analogues on methylation and expression of phosphatase and tensin homologue tumour suppressor gene in breast cancer cells. Br. J. Nutr 107:781–790; 2012. [591] Van Meter, M.; Mao, Z.; Gorbunova, V.; Seluanov, A. Repairing split ends: SIRT6, mono-ADP ribosylation and DNA repair. Aging (Albany NY) 3:829–835; 2011. [592] Chae, H. D.; Broxmeyer, H. E. SIRT1 deficiency downregulates PTEN/JNK/ FOXO1 pathway to block reactive oxygen species-induced apoptosis in mouse embryonic stem cells. Stem Cells Dev. 20:1277–1285; 2011. [593] Janzen, V.; Forkert, R.; Fleming, H. E.; Saito, Y.; Waring, M. T.; Dombkowski, D. M.; Cheng, T.; DePinho, R. A.; Sharpless, N. E.; Scadden, D. T. Stem-cell ageing modified by the cyclin-dependent kinase inhibitor p16INK4a. Nature 443:421–426; 2006. [594] Krishnamurthy, J.; Ramsey, M. R.; Ligon, K. L.; Torrice, C.; Koh, A.; BonnerWeir, S.; Sharpless, N. E. p16INK4a induces an age-dependent decline in islet regenerative potential. Nature 443:453–457; 2006. [595] Molofsky, A. V.; Slutsky, S. G.; Joseph, N. M.; He, S.; Pardal, R.; Krishnamurthy, J.; Sharpless, N. E.; Morrison, S. J. Increasing p16INK4a expression decreases forebrain progenitors and neurogenesis during ageing. Nature 443:448–452; 2006. [596] Liu, S.; Reilly, S. M.; Lee, C. H. Transcriptional repression of mitochondrial function in aging: a novel role for the SMRT co-repressor. Antioxid. Redox Signal. ; August 2, 2012. [Epub ahead of print]. [597] Clark, S. J.; Falchi, M.; Olsson, B.; Jacobson, P.; Cauchi, S.; Balkau, B.; Marre, ˚ M.; Aitman, T. J.; Richardson, S.; M.; Lantieri, O.; Andersson, J. C.; Jernas, ¨ om, ¨ Sjostr L.; Wong, H. Y.; Carlsson, L. M.; Froguel, P.; Walley, A. J. Association of sirtuin 1 (SIRT1) gene SNPs and transcript expression levels with severe obesity. Obesity (Silver Spring) 20:178–185; 2012. [598] Shimoyama, Y.; Suzuki, K.; Hamajima, N.; Niwa, T. Sirtuin 1 gene polymorphisms are associated with body fat and blood pressure in Japanese. Transl. Res. 157:339–347; 2011. [599] Zheng, J.; Chen, L. L.; Xiao, F.; Hu, X.; Deng, X.; Li, H. Three single nucleotide variants of the SIRT1 gene are associated with overweight in a Chinese population: a case control study. Endocr. J. 59:229–237; 2012. [600] Zarrabeitia, M. T.; Valero, C.; Martı´n-Escudero, J. C.; Olmos, J. M.; BoladoCarrancio, A.; de Sande-Nacarino, E. L.; Rodrı´guez-Rey, J. C.; Sainz, J. R. J. A. Association study of Sirtuin 1 polymorphisms with bone mineral density and body mass index. Arch. Med. Res. 43:363–368; 2012. [601] Dong, Y.; Guo, T.; Traurig, M.; Mason, C. C.; Kobes, S.; Perez, J.; Knowler, W. C.; Bogardus, C.; Hanson, R. L.; Baier, L. J. SIRT1 is associated with a decrease in acute insulin secretion and a sex specific increase in risk for type 2 diabetes in Pima Indians. Mol. Genet. Metab. 104:661–665; 2011. [602] Kishi, T.; Yoshimura, R.; Kitajima, T.; Okochi, T.; Okumura, T.; Tsunoka, T.; Yamanouchi, Y.; Kinoshita, Y.; Kawashima, K.; Fukuo, Y.; Naitoh, H.; UmeneNakano, W.; Inada, T.; Nakamura, J.; Ozaki, N.; Iwata, N. SIRT1 gene is associated with major depressive disorder in the Japanese population. J. Affect. Disord. 126:167–173; 2010. [603] Kim, S.; Bi, X.; Czarny-Ratajczak, M.; Dai, J.; Welsh, D. A.; Myers, L.; Welsch, M. A.; Cherry, K. E.; Arnold, J.; Poon, L. W.; Jazwinski, S. M. Telomere maintenance genes SIRT1 and XRCC6 impact age-related decline in telomere length but only SIRT1 is associated with human longevity. Biogerontology 13:119–131; 2012. [604] Flachsbart, F.; Croucher, P. J.; Nikolaus, S.; Hampe, J.; Cordes, C.; Schreiber, S.; Nebel, A. Sirtuin 1 (SIRT1) sequence variation is not associated with exceptional human longevity. Exp. Gerontol. 41:98–102; 2006. [605] Botden, I. P.; Zillikens, M. C.; de Rooij, S. R.; Langendonk, J. G.; Danser, A. H.; Sijbrands, E. J.; Roseboom, T. J. Variants in the SIRT1 gene may affect diabetes risk in interaction with prenatal exposure to famine. Diabetes Care 35:424–426; 2012. [606] Kishi, T.; Fukuo, Y.; Kitajima, T.; Okochi, T.; Yamanouchi, Y.; Kinoshita, Y.; Kawashima, K.; Inada, T.; Kunugi, H.; Kato, T.; Yoshikawa, T.; Ujike, H.; Ozaki, N.; Iwata, N. SIRT1 gene, schizophrenia and bipolar disorder in the Japanese population: an association study. Genes Brain Behav. 10:257–263; 2011. [607] Albani, D.; Polito, L.; Forloni, G. Sirtuins as novel targets for Alzheimer’s disease and other neurodegenerative disorders: experimental and genetic evidence. J. Alzheimers Dis 19:11–26; 2010. [608] Polito, L.; Kehoe, P. G.; Davin, A.; Benussi, L.; Ghidoni, R.; Binetti, G.; Quadri, P.; Lucca, U.; Tettamanti, M.; Clerici, F.; Bagnoli, S.; Galimberti, D.; Nacmias,

[609]

[610]

[611]

[612]

[613]

[614]

[615]

[616]

[617]

[618]

[619] [620]

[621]

[622]

[623]

[624]

[625]

[626]

[627]

171

B.; Sorbi, S.; Guaita, A.; Scarpini, E.; Mariani, C.; Forloni, G.; Albani, D. The SIRT2 polymorphism rs10410544 and risk of Alzheimer’s disease in two Caucasian case-control cohorts. Alzheimers Dement. ; May 30, 2012. [Epub ahead of print]. Rose, G.; Dato, S.; Altomare, K.; Bellizzi, D.; Garasto, S.; Greco, V.; Passarino, G.; Feraco, E.; Mari, V.; Barbi, C.; BonaFe, M.; Franceschi, C.; Tan, Q.; Boiko, S.; Yashin, A. I.; De Benedictis, G. Variability of the SIRT3 gene, human silent information regulator Sir2 homologue, and survivorship in the elderly. Exp. Gerontol. 38:1065–1070; 2003. Bellizzi, D.; Rose, G.; Cavalcante, P.; Covello, G.; Dato, S.; De Rango, F.; Greco, V.; Maggiolini, M.; Feraco, E.; Mari, V.; Franceschi, C.; Passarino, G.; De Benedictis, G. A novel VNTR enhancer within the SIRT3 gene, a human homologue of SIR2, is associated with survival at oldest ages. Genomics 85:258–263; 2005. Lescai, F.; Blanche´, H.; Nebel, A.; Beekman, M.; Sahbatou, M.; Flachsbart, F.; Slagboom, E.; Schreiber, S.; Sorbi, S.; Passarino, G.; Franceschi, C. Human longevity and 11p15.5: a study in 1321 centenarians. Eur. J. Hum. Genet. 17:1515–1519; 2009. Della-Morte, D.; Dong, C.; Bartels, S.; Cabral, D.; Blanton, S. H.; Sacco, R. L.; Rundek, T. Association of the sirtuin and mitochondrial uncoupling protein genes with carotid intima-media thickness. Transl. Res. 160:389–390; 2012. Dong, C.; Della-Morte, D.; Wang, L.; Cabral, D.; Beecham, A.; McClendon, M. S.; Luca, C. C.; Blanton, S. H.; Sacco, R. L.; Rundek, T. Association of the sirtuin and mitochondrial uncoupling protein genes with carotid plaque. PLoS One 6:e27157; 2011. Willcox, B. J.; Donlon, T. A.; He, Q.; Chen, R.; Grove, J. S.; Yano, K.; Masaki, K. H.; Willcox, D. C.; Rodriguez, B.; Curb, J. D. FOXO3A genotype is strongly associated with human longevity. Proc. Natl. Acad. Sci. USA 105:13987–13992; 2008. Donlon, T. A.; Curb, J. D.; He, Q.; Grove, J. S.; Masaki, K. H.; Rodriguez, B.; Elliott, A.; Willcox, D. C.; Willcox, B. J. FOXO3 gene variants and human aging: coding variants may not be key players. J. Gerontol. A Biol. Sci. Med. Sci 67:1132–1139; 2012. ¨ ¨ Flachsbart, F.; Moller, M.; Daumer, C.; Gentschew, L.; Kleindorp, R.; Krawczak, M.; Caliebe, A.; Schreiber, S.; Nebel, A. Genetic investigation of FOXO3A requires special attention due to sequence homology with FOXO3B. Eur. J. Hum Genet. ; May 16, 2012. [Epub ahead of print]. Renault, V. M.; Thekkat, P. U.; Hoang, K. L.; White, J. L.; Brady, C. A.; Kenzelmann Broz, D.; Venturelli, O. S.; Johnson, T. M.; Oskoui, P. R.; Xuan, Z.; Santo, E. E.; Zhang, M. Q.; Vogel, H.; Attardi, L. D.; Brunet, A. The prolongevity gene FoxO3 is a direct target of the p53 tumor suppressor. Oncogene 30:3207–3221; 2011. Kurinna, S.; Stratton, S. A.; Tsai, W. W.; Akdemir, K. C.; Gu, W.; Singh, P.; Goode, T.; Darlington, G. J.; Barton, M. C. Direct activation of forkhead box O3 by tumor suppressors p53 and p73 is disrupted during liver regeneration in mice. Hepatology 52:1023–1032; 2010. Nowak, K.; Killmer, K.; Gessner, C.; Lutz, W. E2F-1 regulates expression of FOXO1 and FOXO3a. Biochim. Biophys. Acta 244-252:2007; 1769. Bakker, W. J.; Harris, I. S.; Mak, T. W. FOXO3a is activated in response to hypoxic stress and inhibits HIF1-induced apoptosis via regulation of CITED2. Mol. Cell 28:941–953; 2007. You, H.; Jang, Y.; You-Ten, A. I.; Okada, H.; Liepa, J.; Wakeham, A.; Zaugg, K.; Mak, T. W. p53-dependent inhibition of FKHRL1 in response to DNA damage through protein kinase SGK1. Proc. Natl. Acad. Sci. USA 101:14057–14062; 2004. Miyaguchi, Y.; Tsuchiya, K.; Sakamoto, K. P53 negatively regulates the transcriptional activity of FOXO3a under oxidative stress. Cell Biol. Int. 33:853–860; 2009. Wang, F.; Marshall, C. B.; Yamamoto, K.; Li, G. Y.; Plevin, M. J.; You, H.; Mak, T. W.; Ikura, M. Biochemical and structural characterization of an intramolecular interaction in FOXO3a and its binding with p53. J. Mol. Biol. 384:590–603; 2008. Zhao, R.; Gish, K.; Murphy, M.; Yin, Y.; Notterman, D.; Hoffman, W. H.; Tom, E.; Mack, D. H.; Levine, A. J. Analysis of p53-regulated gene expression patterns using oligonucleotide arrays. Genes Dev. 14:981–993; 2000. Tran, H.; Brunet, A.; Grenier, J. M.; Datta, S. R.; Fornace, A. J.; DiStefano, P. S.; Chiang, L. W.; Greenberg, M. E. DNA repair pathway stimulated by the forkhead transcription factor FOXO3a through the Gadd45 protein. Science 296:530534; 2002. You, H.; Yamamoto, K.; Mak, T. W. Regulation of transactivationindependent proapoptotic activity of p53 by FOXO3a. Proc. Natl. Acad. Sci. USA 103:9051–9056; 2006. Bouchard, C.; Lee, S.; Paulus-Hock, V.; Loddenkemper, C.; Eilers, M.; Schmitt, C. A. FoxO transcription factors suppress Myc-driven lymphomagenesis via direct activation of Arf. Genes Dev. 21:2775–2787; 2007.